Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2022 May 11;90(5):e00583-21. doi: 10.1128/iai.00583-21

Food as Treatment of Inflammatory Bowel Diseases

Ana Maldonado-Contreras a,
Editor: Karen M Ottemannb
PMCID: PMC9119114  PMID: 35323020

ABSTRACT

Inflammatory bowel diseases (IBD), namely, Crohn’s disease (CD) and ulcerative colitis (UC), are lifelong and incurable chronic inflammatory diseases affecting 6.8 million people worldwide. By 2030, the prevalence of IBD is estimated to reach 1% of the population in Western countries, and thus there is an urgent need to develop effective therapies to reduce the burden of this disease. Microbiome dysbiosis is at the heart of the IBD pathophysiology, and current research and development efforts for IBD treatments have been focused on gut microbiome regulation. Diet can shape the intestinal microbiome. Diet is also preferred over medication, is safe, and has been proven to be an effective strategy for the management of IBD. Therefore, although often overlooked, dietary interventions targeting the microbiome represent ideal treatments for IBD. Here, I summarize the latest research on diet as a treatment for IBD from infancy to adulthood, compile evidence of the mechanisms of action behind diet as treatment, and, lastly, provide insights into future research focusing on culturally tailored diets for ethnic minority groups with increased incidence of IBD yet underrepresented in nutrition research.

KEYWORDS: diet, epithelial barrier, gut inflammation, inflammatory bowel disease, microbiome

INTRODUCTION

The incidence of inflammatory bowel disease (IBD) is on the rise (1). In the United States alone, around 2.5 million people suffer from IBD, resulting in up to $31.6 billion in direct and indirect costs annually (24). Despite the multifactorial causality of IBD, dysbiosis, epithelial barrier dysfunction, and immune disturbances have been suggested as the cornerstones of disease onset and severity (59). Most of the current treatments rely on pharmacological interventions that either dampen inflammation (i.e., corticosteroids, immunosuppressants) or decrease the chance of bacterial breach from the lumen to the underlying mucosa due to a dysfunctional epithelial barrier (i.e., antibiotics). These conventional treatments have remission rates lower than 50% and usually fail to prevent recurrent flare-ups over time. More than one-third of patients with IBD fail induction therapy, and up to 60% of primary responders lose response over time (1012). With a rising prevalence of IBD worldwide (13), there is an urgent need to develop effective and sustainable therapies that can be used long-term.

Interestingly, a microbiome “imbalance” or dysbiosis is linked to barrier dysfunction (1417) and inflammation (1821), which together are responsible for the IBD pathophysiology (8, 22). Over the last decade or so, there has been an increasing enthusiasm about clinical applications of diet as microbiome-targeted therapy as an adjunct treatment of IBD. Diet is a modifiable, noninvasive, inexpensive behavior that is crucial in shaping the intestinal microbiome (2328). Dietary patterns have been associated with increased IBD risk or with the characteristic dysbiosis found in IBD patients in several large cohorts (2933). Thus, diet can serve as a microbiome-targeted adjunct therapy to assist in the management of IBD. To date, intervention studies have shown that diet, in conjunction with medication, is effective in inducing IBD remission and can be tolerated without adverse effects (3445). This review aims to summarize results from studies assessing (i) diet as therapy for IBD, (ii) its effect on the microbiome of patients with IBD, and (iii) the microbiota-dependent mechanisms by which food affects IBD outcomes.

DIETARY TREATMENT FOR IBD ACROSS THE LIFE SPAN

Early life.

Since the pioneering analysis of the Swedish twin registry, mounting evidence demonstrates a robust genetic basis of IBD (46). However, genetics explain only a minority of the variance of disease risk, suggesting that a combination of genetics and environmental factors will be more likely to explain disease pathogenesis. For instance, infants born to mothers with IBD are at a substantially increased risk of developing the disease compared to infants born to fathers with IBD, particularly Crohn’s disease (CD) (47). Akolkar et al. found that out of 135 families where both a parent and a child had IBD, the parent-to-child transmission was linked to the mother in 69% of cases and only 31% of the cases were linked to the father having IBD (48). A second study found a similar trend, with mother-to-child transmissions accounting for 63% of Crohn’s disease cases (49). Finally, studies of children who subsequently developed IBD showed an increased risk (7 to 8 times higher) of Crohn’s disease if the mothers also suffered from the disease (47, 50).

Hence, besides genetics, a possible explanation of the higher proportion of mother-to-child than father-to-child transmissions is that environmental factors during pregnancy might affect the risk of developing IBD. Accumulating evidence suggests that the “inherited” neonate microbiome from the mother can exert marked effects on the immune and metabolic programming of the offspring, with long-term health-related consequences including the predisposition to IBD (5154). Environmental factors during pregnancy, including mode of delivery, perinatal diet, and perinatal antibiotic use, drive the composition of the “inherited” neonate microbiome (55, 56). For instance, infants born by cesarean exhibit a neonate microbiome with low diversity and late colonization of Bacteroidetes that is associated with reduced Th1 cell responses in the first 2 years of life (57). Activation of Th1 triggers the cellular immune response, inhibits macrophage activation, and stimulates B cells to produce IgM and IgG1 antibodies. In addition, experiments in mice have demonstrated that the neonate microbiome can prevent the accumulation of invariant natural killer T cells (iNKT), which results in decreased disease severity in models of IBD (58).

Infants born to mothers with IBD exhibit a higher abundance of Gammaproteobacteria species and depletion of Bifidobacterium species than those born from mothers without IBD (59). Furthermore, maternal IBD status is a significant predictor of the overall β-diversity of the neonate microbiome and of the abundance of bifidobacteria and Gammaproteobacteria over time. Infants born to mothers with IBD had higher levels of fecal calprotectin (FC), an inflammatory marker that robustly correlates with barrier damage and massive neutrophil infiltration in IBD patients (6065). The levels of fecal calprotectin in those babies are higher as early as 2 months and up to 36 months of age than the levels in babies born to healthy mothers (59, 66). Specific bacteria were correlated with fecal calprotectin levels in the infants; namely, Bifidobacterium (depleted in infants born from mothers with IBD), Faecalibacterium, and Alistipes showed negative correlations, and Streptococcus was positively correlated with fecal calprotectin levels within 3 months of birth (66). Experiments with germfree mice inoculated with stools of mothers with IBD showed that mice developed an altered immune maturation. Particularly, mice inoculated with stools of mothers with IBD showed significantly lower levels of switched memory B cells (CD19+ CD27+ IgM IgD) and regulatory T cells (CD3+ CD4+ FoxP3+) than did germfree mice inoculated with stools from pregnant controls (59). The long-term consequences of elevated fecal calprotectin levels or/and an altered immune maturation at an early age are uncertain and hard to evaluate. Nonetheless, restoration of the microbiome in mothers with IBD or their infants can retune the microbiome-dependent immune and metabolic programming of the offspring.

Diet can rapidly and predictably change the microbiome (23). It has been demonstrated that perinatal diet is linked to the “inherited” neonate microbiome (6769). Lundgren et al. analyzed the stool microbiome of 145 infants enrolled in the New Hampshire Birth Cohort Study and compared it to the dietary information obtained during pregnancy from their mothers using a food frequency questionnaire (67). They found that the microbiome of infants born vaginally grouped into three clusters, each cluster dominated by either (i) Bifidobacterium, (ii) Streptococcus and Clostridium, or (iii) Bacteroides. The odds of belonging to the Streptococcus- and Clostridium-dominated cluster was 2.73 times greater for each additional maternal serving of fruit per day. Other foods consumed during pregnancy are associated with specific members of the microbiome in the offspring. For example, high dairy intake during pregnancy was positively associated with Clostridium neonatale, Clostridium butyricum, and Staphylococcus and negatively related to Lachnospiraceae species (67). Another study of mothers in the Spanish-Mediterranean area (n = 116 mothers) showed that the maternal microbiome of this cohort could be grouped into two clusters: Prevotella-dominated cluster I and Ruminococcus-dominated cluster II (69). Mothers in the Ruminococcus-dominated cluster II reported higher intakes of total dietary fiber, monounsaturated omega-3 fatty acids, and polyphenols, while mothers in the Prevotella-dominated cluster I reported higher consumption of carbohydrate and saturated fatty acids (SFA). The authors found that maternal clustering of the microbiome correlated with the neonatal microbiome composition (n = 86 dyads). Infants born from mothers in the Prevotella-dominated cluster I exhibited a higher abundance of Firmicutes spp. than those born from mothers in the Ruminococcus-dominated cluster II. Besides the neonatal microbiome, clustering of the maternal microbiome was also linked to the risk of being overweight. Validated anthropometrical measurements such as weight-for-length (WFL) and body mass index (BMI) Z-scores were collected from infants up to 18 months of age to assess the risk of being overweight. Infants belonging to mothers in the Prevotella-dominated cluster I—with higher consumption of carbohydrate and SAF—and born by cesarean section exhibited significantly higher BMI and WFL Z-scores at 18 months than infants from vaginal births from mothers classified in the Ruminococcus-dominated cluster II—with higher intakes of total dietary fiber, monounsaturated omega-3 fatty acids, and polyphenols (69). Later, Selma-Royo et al. reported a subsequent study including 73 dyads from the same Spanish cohort (68). Here, they showed that intake of SFA and monounsaturated fatty acids (MUFA) during pregnancy significantly affects the overall structure of the offspring microbiome. Particularly, infants of mothers with high SFA and MUFA intakes showed enrichment in Firmicutes (i.e., Coprococcus, Blautia, Roseburia, Ruminococcaceae, and Lachnospiraceae) and depletion in Proteobacteria (i.e., Klebsiella, Escherichia). Conversely, intakes of vegetable-derived proteins and fiber during pregnancy negatively correlate with enrichment of the Firmicutes species mentioned above in the offspring (68).

To the best of my knowledge, only one study in Norway has delved into the dietary patterns of pregnant women with IBD (70). The authors of the study enrolled 183 mothers with CD, 240 mothers with ulcerative colitis (UC), and 83,565 mothers without IBDs. They found that mothers with IBD had low adherence to a “traditional dietary pattern,” characterized by consumption of lean fish, fish products, potatoes, rice porridge, cooked vegetables, and gravy. High adherence to the traditional dietary pattern was associated with improved pregnancy outcomes, namely, a lower risk for “small for gestational age” outcome (70).

My laboratory, in collaboration with researchers at Icahn School of Medicine at Mount Sinai, is currently testing a dietary intervention in mothers with IBD to revert dysbiosis during pregnancy and, in consequence, the “inherited” neonate microbiome and early inflammation seen in infants born to mothers with IBD (45).

Pediatrics.

Exclusive enteral nutrition (EEN) is the primary therapy used to induce remission in pediatric IBD patients, particularly CD (71). This therapy consists of replacing foods with commercial formulas (elemental or polymeric) to provide total calories, complete macronutrients, and micronutrients to pediatric patients with active disease. EEN induces remission in 80 to 85% of pediatric patients, similar to those treated with corticosteroids (72). Compared to steroid treatment, several studies have demonstrated that EEN is superior in inducing mucosal healing in pediatric patients (7377). However, a meta-analysis confirmed that corticosteroid therapy in adults with active disease may be more effective in inducing remission than EEN (78). Thus, EEN is used primarily in pediatric patients.

EEN drives microbiome changes. Specifically, EEN results in a reduction of bacterial diversity along with a decreased abundance of specific short-chain fatty acid (SCFA)-producing bacteria thought to be beneficial (i.e., Faecalibacterium, Ruminoccocus, Blautia, and Subdoligranulum) (79, 80). Moreover, EEN stabilizes the microbiota-dependent metabolism of bile acids (BAs) (80). BAs play a central role in modulating intestinal immune responses and possess antimicrobial activity that can inhibit bacterial overgrowth (81). The liver produces primary BAs from cholesterol, and the gut microbiome can modify these compounds into a myriad of secondary BAs that greatly increase their diversity and biological function (8285). A recent study investigated the BA proportions in fecal samples obtained from 17 CD children before and after EEN treatment. Six of the 17 children did not sustain remission while on EEN treatment and experienced a relapse requiring escalation of medical therapy (e.g., oral corticosteroids, biologic therapy, or surgery). The six children experiencing relapses showed an accumulation of primary BAs in stool before EEN treatment, suggesting depletion of bacteria capable of generating secondary BAs. Those children exhibited a significantly higher abundance of multiple bacteria species unable to modify BAs (i.e., Bacteroides plebeius, Bacteroides ovatus, Bacteroides dorei, Bacteroides thetaiotaomicron, Ruminococcus gnavus, Escherichia coli, Clostridium bolteae, and Veillonella sp.). Conversely, children with EEN-sustained remission (n = 7) exhibited accumulation of microbiome-dependent secondary BAs before EEN and at any given time point (86). Samples from children with EEN-sustained remission showed enrichment of bacterial species with the genetic capacity to modify primary BAs into secondary BAs (i.e., Bacteroides vulgatus, Bacteroides uniformis, Faecalibacterium, Subdoligranulum, and Alistipes sp.) (86). This suggests that microbiota-dependent production of secondary BAs is necessary for EEN-dependent sustained remission.

Other diets tested as a therapy for pediatric patients include the specific carbohydrate diet (SCD) (36, 41), the modified SCD (mSCD) (36), Crohn’s disease treatment with eating (CD-TREAT) (42), and the Crohn’s disease exclusion diet (CDED) (34, 40, 44).

The SCD is one of the most studied diets in the IBD population. SCD focuses on removing grains (i.e., wheat, oats, barley, corn, quinoa, and rice) and milk products (except for hard cheeses and fully fermented yogurts) and replacing any added sugar with honey. Hence, SCD is centered on low complex carbohydrates that can serve as food sources for beneficial bacteria in the colon, thus reverting dysbiosis and reducing inflammation (41). The first study in children with CD (n = 7) showed improvement of inflammatory markers after several months of SCD treatment (41). A few retrospective studies have also confirmed that pediatric patients with CD or UC experienced a reduction of disease activity after SCD treatment (87, 88). Later, a prospective study of children with mild to moderate IBD showed that adding a 12-week treatment with SCD along with concurrent medication induced remission in 8 of the 12 patients included in the study (89). Despite the small sample size and high interpersonal variability, children in the study showed changes in the microbiome, with Bacteroides and Parabacteroides having the largest decrease in median abundance and Eubacterium, Ruminoccocus, and Subdoligranulum the largest increase (89). Of note, depletion of Eubacterium, Ruminoccocus, and Subdoligranulum has been associated with dysbiosis in IBD patients (90). A more recent randomized trial compared the SCD, the mSCD (integrating some initially excluded foods, such as potatoes, rice, quinoa, and oats), and whole foods (eliminating wheat, corn, sugar, milk, and food additives) (36). Here, researchers found that all the children (n = 10) completing 12 weeks on any one of the treatments achieved remission. As expected, children exhibited a microbiome shift that was primarily patient specific (36).

Of note, nutritional deficiencies are common in IBD individuals (i.e., low in calcium, iron, vitamins B6, B9, and B12, vitamin D, and others) (9193). In children with IBD, these deficiencies can lead to growth failure and malnutrition, which are among the major complications of the disease (94). Both EEN and SCD are restrictive diets that are recommended for short-term consumption, as nutritional deficiencies may arise (95, 96).

On that note, investigators in the United Kingdom created a less-restrictive diet for pediatric patients, CD-TREAT (42). CD-TREAT is a diet based on solid food, aimed to recapitulate the nutrient composition and effects of EEN in the microbiome. An initial study on healthy volunteers (n = 25) demonstrated that CD-TREAT is better tolerated than EEN, with similar effects on the microbiome composition and metabolic function (42). Specifically, individuals on either CD-TREAT or EEN showed marked reductions of short-chain fatty acid (SCFA)-producing bacteria, along with lower concentrations of SCFAs in feces. The microbial genetic capacity for BA modification was not assessed in this study. CD-TREAT was then tested in an open-label trial on 5 children with CD who had mild to moderate active luminal disease. Four children completed the study. After 4 weeks on CD-TREAT, 3 patients demonstrated a clinical response (weighted pediatric Crohn’s disease activity index [wPCDAI] score change, >17.5) and 2 were in clinical remission (wPCDAI score, <12.5). At 8 weeks, all the patients showed a clinical response and 3 entered clinical remission (42).

Another whole-food diet, the CDED, was designed to exclude potentially proinflammatory food ingredients, such as gluten, dairy products, gluten-free baked goods and bread, animal fat, processed meats, products containing emulsifiers, canned goods, and all packaged/processed products (44). CDEDs have been tested coupled with partial enteral nutrition (CDED+PEN) but not alone. The first report showed that 33 of 47 children (70%) treated with CDED+PEN for 6 weeks achieved clinical remission (44). In a second study, researchers compared EEN to CDED+PEN and found that the latter is better tolerated by pediatric patients (73.6% versus 97.5%) (34). At week 6, 30 (75%) of 40 children treated with CDED+PEN were in corticosteroid-free remission versus 20 (59%) of 34 children given EEN. The authors assessed the microbiome compositions of 28 pediatric patients on the CDED+PEN and 32 on the EEN. They observed a significant reduction of Actinobacteria and Proteobacteria species and an increased abundance of Clostridia sp. after 6 weeks on either diet. At week 12, patients on the CDED+PEN maintained the dominance of Clostridia and the decrease of Proteobacteria with a minor rebound of Actinobacteria, while patients on the EEN reverted to the pretreatment microbiome composition (34). The latest multicenter randomized trial of CDED+PEN versus EEN in children with mild to moderate CD showed that either treatment resulted in 63% and 67% remission (pediatric Crohn’s disease activity index [PCDAI], <10) rates after 3 and 6 weeks of treatment, respectively (40). See Table 1 for a summary of the trials discussed above.

TABLE 1.

Summary of dietary treatments for IBD tested in human trials

Diet(s) tested (reference) Population tested Study design No. of subjects included Duration of the intervention Outcomesa
EEN (polymeric diet) vs corticosteroids (73) Children with active, naive CD Prospective, randomized, open-label trial n = 37, 19 on the polymeric diet and 18 on corticosteroids 10 wk Remission in 79% of patients on EEN group compared to 67% of patients on corticosteroid group. Mucosa healing was significantly higher in the EEN group, 74%, than in the corticosteriod group, 33%.
EEN (polymeric diet, semi-elemental diet, and elemental diet) vs corticosteroids (74) Children with newly diagnosed active CD Retrospective n = 47, 37 on EEN (12 polymeric; 13 semi-elemental; 12 elemental diet) and 10 on corticosteroids 8 wk Remission in 86.5% patients on EEN vs 90% treated with corticosteroids. Mucosa healing was higher in the EEN group, 64.8%, than in the corticosteroid group, 40%. Compared to group receiving corticosteroids, the duration of clinical remission was longer in the EEN groups, without differences among the three different formulas.
EEN (79) Children with active CD Prospective, nonblinded observational case study with both groups receiving the intervention n = 44, 23 patients with CD and 21 controls 8 wk Remission in 62% of patients with CD. Reduction of bacterial diversity. Reduction of Bifidobacterium, Ruminococcus, and Faecalibacterium.
EEN (80) Children newly diagnosed with CD Prospective, nonblinded observational case study with only CD patients receiving the intervention. Stool sample collected prior to, during, and after EEN. n = 43 6 wk During the intervention, there was a decrease in microbiota diversity and a reduction of amino acids. Also, an increase in microbial metabolism of bile acids. Prior to EEN, microbiota and metabolome are different between responders and nonresponders
SCD (41) Children with CD (PCDAI > 10) Retrospective n = 7 patients with CD 5–30 mo All symptoms resolved 3 mo after SCD. Serum albumin, C-reactive protein, hematocrit, and stool calprotectin either normalized or significantly improved during follow-up clinic visits.
SCD vs mSCD vs whole foods (36) Children with CD, mild to moderate Randomized trial n = 14, 5 on the SCD, 5 on the mSCD, and 4 on the whole food diet. Only 10 completed the study. 12 wk 100% of patients on any of the diets achieved remission. At wk 12, 100% of participants who had elevated CRP at enrollment (n = 8) and completed the study had normal CRP, and 80% (n = 8 out of 10) of participants had a decrease in ESR.
CD-TREAT (42) Children with CD, mild to moderate active luminal disease Prospective, open-label trial n = 5 (only 4 completed the study) 4 wk, with additional 4 wk (follow-up) 3 out of 4 children achieved clinical response, 2 achieved remission (4 wk). All patients achieved clinical response, 3 achieved remission (8 wk).
CDED+PEN or CDED alone (44) Children and young adults with active disease defined by a pediatric Crohn's disease activity index of >7.5 or Harvey-Bradshaw index of ≥4 Prospective, open-label trial n = 47 (7 used CDED without PEN) 6 wk Response and remission were obtained in 37 (78.7%) and 33 (70.2%) patients, respectively. Remission was obtained in 70% of children and 69% of adults. Normalization of previously elevated CRP occurred in 21 of 30 (70%) patients in remission.
CDED+PEN vs EEN (34) Children with CD with short duration of mild to moderate activity, mostly naive to treatment and with small bowel involvement (noninflammatory stricture or resection) Randomized, nonblinded n = 74. Group 1: 40 received CDED + 50% PEN for 6 wk (stage 1), followed by CDED + 25% PEN from wk 7 to 12 (stage 2). Group 2: 34 received EEN in stage 1, followed by a free diet with 25% PEN in stage 2. 12 wk At wk 12, in group 1, 75.5% achieved remission, and of those, 75.9% had a normal CRP, 87.5% sustained remission, and microbiome exhibited dominance of Clostridia and decrease of Proteobacteria. At wk 12, in group 2, 45.1% achieved remission, and of those, 47.6% had a normal CRP, 56% sustained remission, and microbiome reverted to the pretreatment composition. In both groups, fecal calprotectin levels dropped significantly.
SCD vs Mediterranean diet (43) Adult patients with CD, mild to moderate Randomized trial n = 191, 99 on the SCD and 92 on the MD 12 wk At 6 wk, 47% on the SCD and 44% on the MD achieved symptomatic remission with up to 35% showing reduction of fecal calprotectin levels. At wk 12, 42% and 40% on the SCD and MD, respectively, achieved or maintained symptomatic remission. Fecal calprotectin response was observed only in 26% and 8% of patients on the SCD and MD, respectively. No significant change in alpha diversity was observed. In both groups, reduction of Faecalibacterium prausnitzii, Eubacterium eligens, and Eubacterium rectale was detected along with increased abundance of Bacteroides vulgatus and Enterobacteriaceae.
Low FODMAP diet (100) Adults with IBD in remission or with mild disease Randomized trial 6 wk Reduced disease activity and fecal calprotectin along with improvement of self-reported quality of life in patients on low FODMAP diet compared to patients on standard diet.
Low FODMAP vs sham control diet (98) Adults with quiescent IBD Randomized single-blind trial, placebo control trial n = 52 IBD patients, 27 on low FODMAP diet, 25 on placebo diet 4 wk Higher health-related quality of life scores and reduction of symptoms in half of patients on low FODMAP diet compared to control diet. Low abundance of Bifidobacterium adolescentis, Bifidobacterium longum, and Faecalibacterium prausnitzii, along with reduction of total concn of SCFAs in low FODMAP diet-treated patients.
Low-fat, high-fiber diet (LFD) vs improved standard American diet (iSAD) (97) Patients with UC in remission or with mild disease Parallel-group, crossover study. Patients were randomized to an LFD (10% of calories from fat) or an iSAD (35%–40% of calories from fat) for the first 4-wk period, followed by a 2-wk washout period, and then switched to the other diet for 4 wk. n = 17 4 wk on each diet All patients remained in remission throughout the study. Both diets increased self-reported quality of life. Patients showed decreased abundance of Actinobacteria, whereas the relative abundance of Bacteroidetes increased. LFD favored the abundance of Faecalibacterium prausnitzii.
IBD-AID (37) Patients with CD or UC, mild to severe activity Retrospective n = 11 4 wk or more All patients discontinued at least one of their prior IBD medications, and all patients had symptom reduction. Disease activity scores decreased significantly.
IBD-AID (120) Patients with IBD in remission or with mild to severe disease Prospective, open-label trial. First, 6-wk baseline period, followed by an 8-wk intervention period. n = 19 8 wk Consumption of prebiotics, probiotics, and beneficial foods correlated with increased abundance of Clostridia (namely, Roseburia hominis, Faecalibacterium prausnitzii, Eubacterium eligens, Fusicatenibacter saccharivorans) and Bacteroides (namely, Bacteroides dorei, Bacteroides ovatus, and Bacteroides vulgatus) species. Prebiotics and adverse foods have an inverse impact on cytokine levels (i.e., IL-6, IL-8, TNF-α): negatively and positively correlating with proinflammatory cytokines, respectively.
a

CRP, C-reactive protein: ESR, erythrocyte sedimentation rate.

Adults.

“What to eat? is the most frequent question asked by IBD patients of their treating physicians. Only recently has diet been recognized as a cost-effective strategy to induce remission in adult patients with IBD (37, 38, 43, 97). See Table 1 for a summary of the trials discussed below.

A recent randomized trial that included interventions with either the SCD or the Mediterranean diet (MD) has demonstrated a remarkable effect of diet in inducing remission in adults with Crohn’s disease (43). After only 6 weeks, 47% of the patients on the SCD (n = 99) and 44% on the MD (n = 92) achieved symptomatic remission (Crohn’s disease activity index [CDAI], <150), with up to 35% showing reduction in fecal calprotectin (FC) levels (reduction of FC to <250 μg/g and by >50% from screening among those with a screening FC level of >250 μg/g). At week 12, 42% and 40% on the SCD and MD, respectively, achieved or maintained symptomatic remission. The fecal calprotectin response was observed only in 26% and 8% of patients on the SCD and MD, respectively (43). The authors reported no significant changes in microbiome diversity at 0, 6, or 12 weeks post-dietary treatment, with patients in either diet group having comparable richness and Shannon’s diversity indices (43).

The low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (low FODMAP) diet has also been tested to manage irritable bowel syndrome-like symptoms in patients with IBD (38, 98101). A randomized 6-week trial comparing the low FODMAP diet with a standard diet showed that patients (either CD or UC) on the low FODMAP diet modestly reduced their disease activity while those on the standard diet experienced no change in symptoms. Moreover, fecal calprotectin decreased only in the patients adopting the low FODMAP diet (100).

For ulcerative colitis, a catered nutritious low-fat, high-fiber diet (LFD) has been shown to improve the overall quality of life, lower inflammatory markers, and decrease dysbiosis (97). Fritsch et al. carried out a parallel-group crossover study to compare the effect of an LFD (10% of calories from fat) with that of an improved standard American diet (iSAD), which included higher quantities of fruits, vegetables, and fiber than a typical SAD with 35 to 40% of calories from fat. Patients with UC in remission or with mild disease (n = 17) were randomized to either diet for 4 weeks; after a 2-week washout period, patients were switched to the opposite diet. Although there were no significant differences in disease activity scores after each dietary intervention, which were low at baseline (mean partial Mayo score of 0.9), all patients remained in remission during the trial. Based on the validated short IBD questionnaire and the 36-Item Short Form Health Survey scores, participants in either diet group experienced increased quality of life compared to baseline measurements. Inflammatory markers such as serum amyloid A decreased significantly only in participants on the LFD, while microbe-derived metabolite SCFAs, particularly acetate, significantly increased with either dietary treatment. The LFD prompted a significant increase in Bacteroidetes and a decrease in Actinobacteria. In comparison to participants on the iSAD, participants on the LFD showed a significant increase in Faecalibacterium prausnitzi, a potent butyrate-producing bacterium commonly depleted in patients with IBD (90, 102113).

Olendzki et al. have designed the IBD anti-inflammatory diet, or IBD-AID, to revert dysbiosis in IBD patients (37, 45). The IBD-AID promotes the intake of probiotic foods (independent of commercial supplements) to promote the establishment of commensal bacteria, prebiotic foods to feed commensal bacteria, and beneficial foods to meet dietary requirements, and the replacement of adverse foods thought to foster pathogenic microbiota and induce gastrointestinal symptoms (26, 37, 45, 114119). The IBD-AID can be prepared at home, might be healthfully consumed by the entire family long-term, and can be adapted to meet other nutritional concerns as needed. In a retrospective study, both Crohn’s disease (n = 8) and ulcerative colitis (n = 3) patients adopting the IBD-AID experienced a reduction of disease activity and lowered their medication intake after only 4 weeks on the diet. Of the Crohn’s disease patients, the disease activity at baseline measured with the Harvey-Bradshaw index (HBI) averaged 11 (range, 1 to 20), and after dietary intervention, the HBI averaged 1.5 (range, 0 to 3). The ulcerative colitis patients had a mean baseline disease activity score, measured by the modified Truelove and Witts severity index (MTLWSI), of 7 (range 6 to 8), and their mean follow-up score was 0. The mean decrease in the HBI was 9.5, and the mean decrease in the MTLWSI was 7 (37). After the IBD-AID intervention, patients exhibited an increased abundance of potent butyrate-producing Faecalibacterium prausnitzii, Eubacterium eligens, and Roseburia hominis (120), all of which are commonly depleted in patients with IBD (90, 102113).

MOLECULAR MECHANISM OF DIET-MICROBIOME INTERACTION LEADING TO AMELIORATION OF IBD

The precise knowledge of the mechanisms by which food affects IBD will catalyze personalized nutritional therapy, sensitivity to differences in IBD clinical manifestations, host genotype, gut microbiome composition, and genetic capabilities. Compiling in vitro and in vivo evidence has shed light on the mechanisms by which diet decreases manifestations of the disease. A recent review summarizes studies with models of diseases detailing mechanistic findings (121). Here, I focus on the diet-microbiome interactions that affect the epithelial barrier permeability and the immune response in IBD.

Dysbiosis and the epithelial barrier.

Dysbiosis in patients with IBD features a depletion of the commensal Clostridia and an expansion of Enterobacteriaceae species known to impact barrier function (90, 103113, 122125). Disruption of the epithelial barrier in IBD leads to the breach of bacteria and foreign antigens from the lumen into the underlying mucosa (8, 126128). Once the epithelial barrier is infringed, a potent inflammatory response is activated, furthering the epithelial damage (129). Barrier dysfunction precedes IBD onset (130139). Moreover, intestinal permeability is a robust predictor of poor outcomes and disease recurrence (132, 140143). The gut microbiome is crucial in supporting a functioning epithelial barrier. For instance, the microbiome-derived SCFAs acetate, propionate, and butyrate represent the primary energy source for enterocytes (144), can act on genes involved in tight junction to seal the paracellular space (14, 145147), increase oxygen consumption in the intestinal epithelium, which in turn stabilizes the hypoxia-inducible factor (HIF), a transcription factor responsible for maintaining barrier integrity (148), and sets an anti-inflammatory tone in the gut mucosa (1417).

Plant-based foods, rich in fiber, are linked to increased abundance of SCFA-producing bacteria in IBD patients (32, 149). Conversely, fiber-deprived diets can cause a dysfunctional epithelial barrier. Schroeder et al. showed that mice fed a Western-like diet (high-fat/low-fiber) have an increased mucosal permeability and a reduced growth rate of the mucus layer (150). Transplantation of the microbiome of mice fed with chow into the mice fed a Western-like diet restores mucosal permeability and mucus growth. This highlights the importance of diet-dependent changes of the microbiome in mucosal barrier integrity and permeability (150). In a separate study, a low-fiber diet caused mucosal epithelial erosion, which in turn promoted lethal fulminant colitis in mice (151). Mice on low-fiber diets showed an expansion of mucus-foraging bacteria, such as Akkermansia muciniphila (152) and Bacteroides caccae (153), in comparison to mice on fiber-rich diets. The expansion of these mucolytic bacteria resulted in a permeable, eroded, thinner epithelial layer, with mice exhibiting intestinal inflammation (i.e., neutrophil infiltration, shorter colon length) and increased susceptibility to Citrobacter-induced colitis (151).

A high-fat diet also impairs barrier function, leading to susceptibility to colitis. A study from Xie et al. demonstrated that the 3-week offspring of mice fed a high-fat diet during pregnancy and lactation exhibited an abnormal epithelial layer with shorter villi, decreased crypt depth, and reduced number of proliferating cells, which in turn led to a lower number of differentiated intestinal cells, in comparison to animals fed a low-fat control diet (154). These morphological defects were accompanied by increased barrier permeability (measured by fluorescein isothiocyanate [FITC] in the serum) and decreased expression of tight junction proteins claudin 1 (CLDN), CLDN3, ZO-1, and occludin. Similarly, patients with severe IBD exhibit a compromised epithelial barrier with low levels of tight junction proteins, specifically occludin (155158).

Comparisons of the gut microbiome of the offspring also revealed striking differences. Offspring of mice fed a high-fat diet had a lower alpha diversity than controls, an increased abundance of Akkermansia muciniphila, Peptostreptococcaceae, and Streptococcus, and a decreased abundance of butyrate-producing bacteria such as Lachnospiraceae incertae sedis and Prevotellaceae. Not surprisingly, the offspring of mice fed a high-fat diet and treated with dextran sodium sulfate (DSS; a sulfated polysaccharide widely used to reproducibly induce experimental acute and chronic colitis) exhibited more severe colitis than their counterparts (154). Similarly, adult mice fed a high-fat diet exhibited a significantly reduced expression of the tight junction protein occludin, which in turn compromised the epithelial barrier, leading to translocation of endotoxin (159).

In an elegant animal study, researchers tested the effect of over 40 diets, with various concentrations, combinations, and sources of macronutrients, on the microbiome, intestinal permeability, and colitis severity (160). They found that mice on diets high in protein (sources included chicken, beef, and egg whites) had increased intestinal permeability, weight loss, and severe colitis than mice on high-fiber diets. The effect of a high-protein diet (particularly casein) on colitis severity was mediated by the gut microbiome, since a high-casein diet reduces survival in comparison to a low-casein diet in specific-pathogen-free (SPF) mice but not in germfree animals. Moreover, casein-driven changes in gut microbial density were significantly associated with the severity of colitis seen in mice. Conversely, fiber-rich diets (particularly, the soluble fiber psyllium) increased the survival of mice treated with DSS (by at least 15 days compared to those on high-protein diets) and reduced colitis severity and the disruption of the epithelial permeability. The effects of dietary psyllium on colitis severity were both dependent and independent of the microbiome (160).

Another important food component to consider is dietary emulsifiers (i.e., carboxymethylcellulose, polysorbate 80, carrageenan, etc.). Emulsifiers are a ubiquitous component of processed food with detrimental effects on barrier function (161164). Mice with chronic exposure to dietary emulsifiers exhibit erosion of the intestinal mucus layer and, in consequence, an enrichment of mucosa-associated inflammation-promoting Proteobacteria species (162, 163). More recently, a study showed that emulsifiers not only alter the mucosa-associated microbiota but also directly induce the expression of bacterial virulence genes to trigger chronic inflammation in mice (165).

In sum, evidence in animal models supports the importance of fiber-rich/low-fat/low-protein, emulsifier-restrictive diets to strengthen epithelial barrier function, which can contribute to resistance to colitis.

Dysbiosis and the immune response.

The immune signature of IBD features an exacerbated epithelial infiltration of innate immune cells (i.e., neutrophils, macrophages, and dendritic cells) accompanied by an excessive activation of effector T cells (Th1, Th2, Th17, and T follicular helper [Tfh] cells) and/or altered tolerance mechanisms mediated by regulatory T cells (Tregs). Particularly, forkhead box protein P3 (FOXP3+) regulatory T cells or FOXP3+ Treg cells, located in the gut lamina propria, function as key regulators of intestinal inflammation in IBD (166170). It has been established that Clostridia species, missing in IBD patients, are responsible for the activation of potent anti-inflammatory FOXP3+ Treg cells (18, 171, 172). Although there is an overall increase of Clostridia in patients adopting some of the IBD-friendly diets aforementioned, the impact of the diet-dependent Clostridia enrichment on Treg activation has not been studied in IBD patients undergoing dietary treatment. Yet, this provides a plausible hypothesis as one of the mechanisms behind those diets.

In a recent study, Song et al. aimed to investigate the diet-microbiome interactions that lead to the induction of Tregs (173). They compared induction of Tregs in three groups of mice: (i) specific-pathogen-free (SPF) mice fed a nutrient-rich diet, (ii) SPF mice fed a minimal diet, and (iii) germfree mice fed a nutrient-rich diet (173). Lower induction of Tregs was observed in SPF mice fed a minimal diet and germfree mice on a nutrient-rich diet, supporting the notion that both a rich diet and a functional microbiome are necessary for Treg induction. Moreover, investigators demonstrated that specific combinations of murine primary and secondary BAs added to the drinking water of SPF mice fed a minimal diet and germfree mice on a nutrient-rich diet restored Treg induction comparable to that of SPF mice fed a nutrient-rich diet. Deletion of either gene involved in BA deconjugation (bile salt hydrolase [BSH]) or the entire BA metabolic pathway in Bacteroides reduced the Treg induction. When susceptibility to colitis was tested, SPF mice on the minimal diet—with a lower proportion of colonic Tregs—exhibited higher weight loss and more severe colitis than SPF mice fed a rich diet. Although supplementation of primary and secondary BAs increased Treg cell counts in SPF mice on the minimal diet, after colitis onset, BA supplementation did not improve colitis in these mice. This highlights the importance of an initial BA-activated Treg pool to confer resistance to chemically induced colitis (173).

Devkota et al. found that mice on a diet high in saturated milk fat promoted taurine conjugation of BAs in the liver, which in turn increased the availability of organic sulfur that can be used by the taurine-respiring sulfidogenic organism Bilophila wadsworthia (174). B. wadsworthia is considered a pathobiont with a high capacity to reduce sulfites. The expansion of this bacterium was associated with a proinflammatory TH1 immune response and with a greater number of genetically susceptible IL-10−/− mice developing colitis. In support of these findings, data from human subjects have shown that consumption of highly saturated fats (mainly animal fat) significantly increases taurine conjugation of BAs, production of fecal sulfide, and abundance of B. wadsworthia (23, 175177).

Similarly, Sinha et al. demonstrated that microbiome-dependent BA modification causes intestinal inflammation in murine models of colitis (178). Here, investigators performed metabolomic analyses of stools from ulcerative colitis-pouch patients and found that there was a reduction in secondary BAs in ulcerative colitis-pouch patients, namely, lithocholic acid (LCA) and deoxycholic acid (DCA), while the accumulation of primary BAs was significantly higher than that in a control group. This metabolic profile was accompanied by a reduction of Ruminococcaceae sp. (involved in secondary BA production [179, 180]). Mice supplemented with secondary BAs showed a significant decrease in chemokines and cytokines, linked to inflammation, and an overall reduction of intestinal inflammation. The authors tested a chemically induced model of colitis (DSS and 2,4,6-trinitrobenzenesulfonic acid [TNBS]) as well as T cell transfer (CD45RBhi) murine models of colitis supplemented with the secondary BAs LCA and DCA. In every model of colitis, they observed a reduction of colitis as measured by weight loss, colon length, gross colon morphology, leukocyte infiltration, histology, and fecal lipocalin 2 when treated with secondary BAs. This effect was abrogated in mice lacking the G protein-coupled receptor TGR5, a transmembrane BA receptor. It has been demonstrated that secondary BA TGR5 agonists, DCA and LCA, can inhibit the production of tumor necrosis factor alpha (TNF-α) in CD14+ macrophages (181). Of interest, TNF-α is a proinflammatory mediator that plays an integral role in the pathogenesis of IBD.

Microbiome-derived SCFAs, favored by fiber-rich diets, also play a fundamental role in mediating the immune tone. In mice, fiber intake during pregnancy increases levels of butyrate in the blood of the offspring with concomitant increased numbers of peripheral and thymic Treg (182). Moreover, it has been shown that EEN supplemented with a multifiber mix prompted an expansion of mucosal CD4+ Foxp3+ Tregs along with an increase in concentrations of total SCFAs, i.e., acetate, propionate, and butyrate, in colitis-susceptible mice (IL-10−/−) (183). In addition, fiber supplementation reduced disease pathology and restored barrier function in IL-10−/− mice (183). In contrast, mice on a high-fat diet exhibited reduced thymocyte counts and increased apoptosis of developing T cell populations (184). Butyrate can also activate Treg function. By acting on Treg, microbiome-derived butyrate reduces levels of proinflammatory cytokines, including TNF-α, interleukin 6 (IL-6), IL-1β, and MCP1/CCL2 (185187). Moreover, microbe-derived butyrate promotes monocyte-to-macrophage differentiation via histone deacetylase inhibition. The macrophages differentiated by the addition of butyrate showed an enhanced antimicrobial activity, which in vivo increased colonization resistance to enteropathogens (188).

In vitro studies have demonstrated that other SCFAs, specifically acetate and propionate, promote differentiation of naive CD4+ T cells into Th17 cells with concomitant induction of IL-17A, IL-17F, RORα, RORγt, T-bet, and IFN-γ (189, 190). These SCFA effects on T cells combine histone deacetylase inhibitor-dependent and -independent mechanisms (189, 190).

In sum, increased evidence points at the pivotal role of diet-dependent changes of the microbiome in the immune response leading to/preventing colitis.

FUTURE CONSIDERATIONS: CULTURALLY TAILORED DIETARY INTERVENTION FOR UNDERREPRESENTED IBD PATIENTS

Historically, IBD is known to affect more people of Caucasian origin than other ethnic groups; however, there is emerging evidence that the prevalence of IBD in Hispanics may be increasing, along with that in the general U.S. population (191, 192). Currently, Hispanics and Latinx account for over 18% of the U.S. population (193, 194). Foreign-born Hispanics in the United States are diagnosed at an older age and present more cases of ulcerative colitis than U.S.-born Hispanics and non-Hispanic whites (195). A meta-analysis also showed that Crohn’s disease behavior between non-Hispanic whites and Hispanics is similar, but Hispanics had a tendency of less upper gastrointestinal involvement (196). Growing evidence demonstrates that Hispanics change their diet upon immigration to the United States, reporting low consumption of total vegetables, legumes, whole grains, and sea plant protein (197). Similarly, Asians, the fastest-growing racial group in the United States (194), have been increasingly diagnosed with IBD in the United States and around the world (198, 199). Recent studies have demonstrated that Asians exhibit different IBD clinical phenotypes, including ocular manifestations and more fistulizing perianal Crohn’s disease, than their Caucasian counterparts (198, 199). A comprehensive study confirmed that U.S. immigrants (of Asian origins) suffer an immediate loss of gut microbiome diversity along with a reduction of microbial capacity for fiber degradation (200). Instead, the microbiome of Asian immigrants is characterized by an enrichment of United States-associated bacterial strains displacing native strains along with the genetic capabilities (200). Like Hispanics, Asian immigrants rapidly change their diet upon arrival in the United States, which partially explains the shift in the microbiome seen in this population (200).

The lack of inclusion of underrepresented ethnic minorities in IBD studies has ignited efforts led by patient advocates and IBD specialists of South Asian descent, such as the South Asian IBD Alliance (SAIAI) (201). The alliance aims to promote “the need for culturally competent, evidence-based, patient-centric care via advocacy, education, and training” to improve the care of South Asian patients with IBD across the globe (201).

Despite the increased incidence of IBD in these minority groups, research aimed at these populations, including research on diet as therapy for IBD, is lacking. In addition, ethnic minority groups frequently experience low-quality care at hospitals due to a combination of factors, including lack of insurance, economic and language barriers, and racial bias in pain assessment and treatment recommendations, to name a few (202204). Culturally tailored interventions can close the gap in the paucity of research and help improve health care equity and quality for minority populations with IBD (205208).

Culturally tailored interventions are frequently implemented in the context of behavioral health trials, with proven success to encourage healthy behaviors (including healthy eating) and to address health disparities affecting minority groups with chronic diseases (206, 209211). A recent meta-analysis of 33 culturally tailored trials highlighted three key aspects of successful interventions (212), as follows. (i) “Linguistic tailoring” aims to address not only the language but also the literacy needs of the target population. Moreover, linguistic tailoring should also consider the inclusion of bilingual staff to remove language barriers between patients/participants, research staff, and health care providers. (ii) “Sociocultural tailoring” aims to incorporate cultural values, unique experiences, religious beliefs, and behaviors of the target group. (iii) “Constituent-involving strategies” aims to build on a sense of collectivism and existing kinship networks by including members of the target community in the research and intervention activities, from actively participating in the study design to their involvement in delivering the intervention (212, 213).

In the case of nutrition, culturally tailored interventions also need to be adapted to the unique culinary preferences and access to foods of the target community. By doing so, the interventions will be relevant to understudied minority groups with high IBD prevalence and in need of attainable strategies to improve their quality of life.

Another challenge for culturally tailored dietary interventions is long-term compliance. Examples from data about dietary recommendations in Australia (214) demonstrate that it is not enough to solely suggest that people consume more beneficial foods. Therefore, culturally tailored dietary interventions also need to provide patients with culinary training aimed at building skills and confidence in food preparation in the kitchen (215).

ACKNOWLEDGMENTS

Ana Maldonado-Contreras is supported by the American Gastroenterological Association and the Leona M. and Harry B. Helmsley Charitable Trust.

Biography

graphic file with name iai.00583-21-f001.gif

Ana Maldonado-Contreras received her master’s degree in microbiology at the Instituto Venezolano de Investigaciones Científicas and completed her Ph.D. at the University of Puerto Rico. She is currently an Assistant Professor at the University of Massachusetts Chan Medical School. Over the last 15 years, Dr. Maldonado-Contreras' research has focused on pathogens and commensal bacteria residing in the gastrointestinal epithelium. For her graduate work, she demonstrated the role of Helicobacter pylori in shaping the gastric microbiota community. Later, she investigated the cross talk between enteropathogens and the intestinal epithelium to identify key mediators of epithelial barrier function and the immune response during bacterial infection. Her current work focuses on the understanding of microbiome-centered therapy (i.e., diet) and its impact on epithelial barrier function and host immune networks.

Contributor Information

Ana Maldonado-Contreras, Email: Ana.Maldonado@umassmed.edu.

Karen M. Ottemann, University of California, Santa Cruz

REFERENCES

  • 1.GBD 2017 Inflammatory Bowel Disease Collaborators. 2020. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 5:17–30. doi: 10.1016/S2468-1253(19)30333-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.CCFA. 2014. Crohn’s & Colitis Foundation of America: the facts about inflammatory bowel diseases. https://www.crohnscolitisfoundation.org/sites/default/files/2019-02/Updated%20IBD%20Factbook.pdf.
  • 3.Kappelman MD, Moore KR, Allen JK, Cook SF. 2013. Recent trends in the prevalence of Crohn's disease and ulcerative colitis in a commercially insured US population. Dig Dis Sci 58:519–525. doi: 10.1007/s10620-012-2371-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Longobardi T, Jacobs P, Bernstein CN. 2003. Work losses related to inflammatory bowel disease in the United States: results from the National Health Interview Survey. Am J Gastroenterol 98:1064–1072. doi: 10.1111/j.1572-0241.2003.07285.x. [DOI] [PubMed] [Google Scholar]
  • 5.Dupaul-Chicoine J, Dagenais M, Saleh M. 2013. Crosstalk between the intestinal microbiota and the innate immune system in intestinal homeostasis and inflammatory bowel disease. Inflamm Bowel Dis 19:2227–2237. doi: 10.1097/MIB.0b013e31828dcac7. [DOI] [PubMed] [Google Scholar]
  • 6.Podolsky DK. 2003. The future of IBD treatment. J Gastroenterol 38(Suppl 15):63–66. [PubMed] [Google Scholar]
  • 7.Tamboli CP, Neut C, Desreumaux P, Colombel JF. 2004. Dysbiosis in inflammatory bowel disease. Gut 53:1–4. doi: 10.1136/gut.53.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Martini E, Krug SM, Siegmund B, Neurath MF, Becker C. 2017. Mend your fences: the epithelial barrier and its relationship with mucosal immunity in inflammatory bowel disease. Cell Mol Gastroenterol Hepatol 4:33–46. doi: 10.1016/j.jcmgh.2017.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vindigni SM, Zisman TL, Suskind DL, Damman CJ. 2016. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: a tripartite pathophysiological circuit with implications for new therapeutic directions. Therap Adv Gastroenterol 9:606–625. doi: 10.1177/1756283X16644242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Peyrin-Biroulet L, Lemann M. 2011. Review article: remission rates achievable by current therapies for inflammatory bowel disease. Aliment Pharmacol Ther 33:870–879. doi: 10.1111/j.1365-2036.2011.04599.x. [DOI] [PubMed] [Google Scholar]
  • 11.Martinez-Montiel MP, Casis-Herce B, Gomez-Gomez GJ, Masedo-Gonzalez A, Yela-San Bernardino C, Piedracoba C, Castellano-Tortajada G. 2015. Pharmacologic therapy for inflammatory bowel disease refractory to steroids. Clin Exp Gastroenterol 8:257–269. doi: 10.2147/CEG.S58152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ahmed W, Galati J, Kumar A, Christos PJ, Longman R, Lukin DJ, Scherl E, Battat R. 2021. Dual biologic or small molecule therapy for treatment of inflammatory bowel disease: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 20:e361–e379. doi: 10.1016/j.cgh.2021.03.034. [DOI] [PubMed] [Google Scholar]
  • 13.Kaplan GG, Windsor JW. 2021. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 18:56–66. doi: 10.1038/s41575-020-00360-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wang RX, Lee JS, Campbell EL, Colgan SP. 2020. Microbiota-derived butyrate dynamically regulates intestinal homeostasis through regulation of actin-associated protein synaptopodin. Proc Natl Acad Sci USA 117:11648–11657. doi: 10.1073/pnas.1917597117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Geirnaert A, Calatayud M, Grootaert C, Laukens D, Devriese S, Smagghe G, De Vos M, Boon N, Van de Wiele T. 2017. Butyrate-producing bacteria supplemented in vitro to Crohn's disease patient microbiota increased butyrate production and enhanced intestinal epithelial barrier integrity. Sci Rep 7:11450. doi: 10.1038/s41598-017-11734-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Segain JP, Raingeard de la Bletiere D, Bourreille A, Leray V, Gervois N, Rosales C, Ferrier L, Bonnet C, Blottiere HM, Galmiche JP. 2000. Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn's disease. Gut 47:397–403. doi: 10.1136/gut.47.3.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chang PV, Hao L, Offermanns S, Medzhitov R. 2014. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci USA 111:2247–2252. doi: 10.1073/pnas.1322269111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B, Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K. 2013. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500:232–236. doi: 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
  • 19.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S, Ivanov II, Umesaki Y, Itoh K, Honda K. 2011. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331:337–341. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Brown K, DeCoffe D, Molcan E, Gibson DL. 2012. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients 4:1095–1119. doi: 10.3390/nu4081095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cavicchia PP, Steck SE, Hurley TG, Hussey JR, Ma Y, Ockene IS, Hébert JR. 2009. A new dietary inflammatory index predicts interval changes in serum high-sensitivity C-reactive protein. J Nutr 139:2365–2372. doi: 10.3945/jn.109.114025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Clayburgh DR, Barrett TA, Tang Y, Meddings JB, Van Eldik LJ, Watterson DM, Clarke LL, Mrsny RJ, Turner JR. 2005. Epithelial myosin light chain kinase-dependent barrier dysfunction mediates T cell activation-induced diarrhea in vivo. J Clin Invest 115:2702–2715. doi: 10.1172/JCI24970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. 2014. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505:559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. 2010. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA 107:14691–14696. doi: 10.1073/pnas.1005963107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. 2009. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 1:6ra14. doi: 10.1126/scitranslmed.3000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ang QY, Alexander M, Newman JC, Tian Y, Cai J, Upadhyay V, Turnbaugh JA, Verdin E, Hall KD, Leibel RL, Ravussin E, Rosenbaum M, Patterson AD, Turnbaugh PJ. 2020. Ketogenic diets alter the gut microbiome resulting in decreased intestinal Th17 cells. Cell 181:1263–1275.e16. doi: 10.1016/j.cell.2020.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.De Angelis M, Ferrocino I, Calabrese FM, De Filippis F, Cavallo N, Siragusa S, Rampelli S, Di Cagno R, Rantsiou K, Vannini L, Pellegrini N, Lazzi C, Turroni S, Lorusso N, Ventura M, Chieppa M, Neviani E, Brigidi P, O'Toole PW, Ercolini D, Gobbetti M, Cocolin L. 2020. Diet influences the functions of the human intestinal microbiome. Sci Rep 10:4247. doi: 10.1038/s41598-020-61192-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zmora N, Suez J, Elinav E. 2019. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol 16:35–56. doi: 10.1038/s41575-018-0061-2. [DOI] [PubMed] [Google Scholar]
  • 29.Lee D, Albenberg L, Compher C, Baldassano R, Piccoli D, Lewis JD, Wu GD. 2015. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology 148:1087–1106. doi: 10.1053/j.gastro.2015.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tjonneland A, Overvad K, Bergmann MM, Nagel G, Linseisen J, Hallmans G, Palmqvist R, Sjodin H, Hagglund G, Berglund G, Lindgren S, Grip O, Palli D, Day NE, Khaw KT, Bingham S, Riboli E, Kennedy H, Hart A, IBD in EPIC Study Investigators. 2009. Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case-control study within a European prospective cohort study. Gut 58:1606–1611. doi: 10.1136/gut.2008.169078. [DOI] [PubMed] [Google Scholar]
  • 31.Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, de Silva P, Fuchs CS, Willett WC, Richter JM, Chan AT. 2014. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn's disease. Gut 63:776–784. doi: 10.1136/gutjnl-2013-305304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bolte LA, Vich Vila A, Imhann F, Collij V, Gacesa R, Peters V, Wijmenga C, Kurilshikov A, Campmans-Kuijpers MJE, Fu J, Dijkstra G, Zhernakova A, Weersma RK. 2021. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut 70:1287–1298. doi: 10.1136/gutjnl-2020-322670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Narula N, Wong ECL, Dehghan M, Mente A, Rangarajan S, Lanas F, Lopez-Jaramillo P, Rohatgi P, Lakshmi PVM, Varma RP, Orlandini A, Avezum A, Wielgosz A, Poirier P, Almadi MA, Altuntas Y, Ng KK, Chifamba J, Yeates K, Puoane T, Khatib R, Yusuf R, Bostrom KB, Zatonska K, Iqbal R, Weida L, Yibing Z, Sidong L, Dans A, Yusufali A, Mohammadifard N, Marshall JK, Moayyedi P, Reinisch W, Yusuf S. 2021. Association of ultra-processed food intake with risk of inflammatory bowel disease: prospective cohort study. BMJ 374:n1554. doi: 10.1136/bmj.n1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Levine A, Wine E, Assa A, Sigall Boneh R, Shaoul R, Kori M, Cohen S, Peleg S, Shamaly H, On A, Millman P, Abramas L, Ziv-Baran T, Grant S, Abitbol G, Dunn KA, Bielawski JP, Van Limbergen J. 2019. Crohn's disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology 157:440–450.e8. doi: 10.1053/j.gastro.2019.04.021. [DOI] [PubMed] [Google Scholar]
  • 35.Luo Y, Yu J, Lou J, Fang Y, Chen J. 2017. Exclusive enteral nutrition versus infliximab in inducing therapy of pediatric Crohn's disease. Gastroenterol Res Pract 2017:6595048. doi: 10.1155/2017/6595048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Suskind DL, Lee D, Kim YM, Wahbeh G, Singh N, Braly K, Nuding M, Nicora CD, Purvine SO, Lipton MS, Jansson JK, Nelson WC. 2020. The specific carbohydrate diet and diet modification as induction therapy for pediatric Crohn's disease: a randomized diet controlled trial. Nutrients 12:3749. doi: 10.3390/nu12123749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Olendzki BC, Silverstein TD, Persuitte GM, Ma Y, Baldwin KR, Cave D. 2014. An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report. Nutr J 13:5. doi: 10.1186/1475-2891-13-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Prince AC, Myers CE, Joyce T, Irving P, Lomer M, Whelan K. 2016. Fermentable carbohydrate restriction (low FODMAP diet) in clinical practice improves functional gastrointestinal symptoms in patients with inflammatory bowel disease. Inflamm Bowel Dis 22:1129–1136. doi: 10.1097/MIB.0000000000000708. [DOI] [PubMed] [Google Scholar]
  • 39.Sigall Boneh R, Sarbagili Shabat C, Yanai H, Chermesh I, Ben Avraham S, Boaz M, Levine A. 2017. Dietary therapy with the Crohn's disease exclusion diet is a successful strategy for induction of remission in children and adults failing biological therapy. J Crohns Colitis 11:1205–1212. doi: 10.1093/ecco-jcc/jjx071. [DOI] [PubMed] [Google Scholar]
  • 40.Sigall Boneh R, Van Limbergen J, Wine E, Assa A, Shaoul R, Milman P, Cohen S, Kori M, Peleg S, On A, Shamaly H, Abramas L, Levine A. 2021. Dietary therapies induce rapid response and remission in pediatric patients with active Crohn's disease. Clin Gastroenterol Hepatol 19:752–759. doi: 10.1016/j.cgh.2020.04.006. [DOI] [PubMed] [Google Scholar]
  • 41.Suskind DL, Wahbeh G, Gregory N, Vendettuoli H, Christie D. 2014. Nutritional therapy in pediatric Crohn disease: the specific carbohydrate diet. J Pediatr Gastroenterol Nutr 58:87–91. doi: 10.1097/MPG.0000000000000103. [DOI] [PubMed] [Google Scholar]
  • 42.Svolos V, Hansen R, Nichols B, Quince C, Ijaz UZ, Papadopoulou RT, Edwards CA, Watson D, Alghamdi A, Brejnrod A, Ansalone C, Duncan H, Gervais L, Tayler R, Salmond J, Bolognini D, Klopfleisch R, Gaya DR, Milling S, Russell RK, Gerasimidis K. 2019. Treatment of active Crohn's disease with an ordinary food-based diet that replicates exclusive enteral nutrition. Gastroenterology 156:1354–1367.e6. doi: 10.1053/j.gastro.2018.12.002. [DOI] [PubMed] [Google Scholar]
  • 43.Lewis JD, Sandler RS, Brotherton C, Brensinger C, Li H, Kappelman MD, Daniel SG, Bittinger K, Albenberg L, Valentine JF, Hanson JS, Suskind DL, Meyer A, Compher CW, Bewtra M, Saxena A, Dobes A, Cohen BL, Flynn AD, Fischer M, Saha S, Swaminath A, Yacyshyn B, Scherl E, Horst S, Curtis JR, Braly K, Nessel L, McCauley M, McKeever L, Herfarth H, DINE-CD Study Group. 2021. A randomized trial comparing the specific carbohydrate diet to a Mediterranean diet in adults with Crohn’s disease. Gastroenterology 161:837–852.e9. doi: 10.1053/j.gastro.2021.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sigall-Boneh R, Pfeffer-Gik T, Segal I, Zangen T, Boaz M, Levine A. 2014. Partial enteral nutrition with a Crohn's disease exclusion diet is effective for induction of remission in children and young adults with Crohn's disease. Inflamm Bowel Dis 20:1353–1360. doi: 10.1097/MIB.0000000000000110. [DOI] [PubMed] [Google Scholar]
  • 45.Peter I, Maldonado-Contreras A, Eisele C, Frisard C, Simpson S, Nair N, Rendon A, Hawkins K, Cawley C, Debebe A, Tarassishin L, White S, Dubinsky M, Stone J, Clemente JC, Sabino J, Torres J, Hu J, Colombel JF, Olendzki B. 2020. A dietary intervention to improve the microbiome composition of pregnant women with Crohn's disease and their offspring: the MELODY (Modulating Early Life Microbiome through Dietary Intervention in Pregnancy) trial design. Contemp Clin Trials Commun 18:100573. doi: 10.1016/j.conctc.2020.100573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tysk C, Lindberg E, Jarnerot G, Floderus-Myrhed B. 1988. Ulcerative colitis and Crohn's disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut 29:990–996. doi: 10.1136/gut.29.7.990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jolving LR, Nielsen J, Beck-Nielsen SS, Nielsen RG, Friedman S, Kesmodel US, Norgard BM. 2017. The association between maternal chronic inflammatory bowel disease and long-term health outcomes in children—a nationwide cohort study. Inflamm Bowel Dis 23:1440–1446. doi: 10.1097/MIB.0000000000001146. [DOI] [PubMed] [Google Scholar]
  • 48.Akolkar PN, Gulwani-Akolkar B, Heresbach D, Lin XY, Fisher S, Katz S, Silver J. 1997. Differences in risk of Crohn's disease in offspring of mothers and fathers with inflammatory bowel disease. Am J Gastroenterol 92:2241–2244. [PubMed] [Google Scholar]
  • 49.Zelinkova Z, Stokkers PC, van der Linde K, Kuipers EJ, Peppelenbosch MP, van der Woude CP. 2012. Maternal imprinting and female predominance in familial Crohn's disease. J Crohns Colitis 6:771–776. doi: 10.1016/j.crohns.2012.01.002. [DOI] [PubMed] [Google Scholar]
  • 50.Roberts SE, Wotton CJ, Williams JG, Griffith M, Goldacre MJ. 2011. Perinatal and early life risk factors for inflammatory bowel disease. World J Gastroenterol 17:743–749. doi: 10.3748/wjg.v17.i6.743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gensollen T, Iyer SS, Kasper DL, Blumberg RS. 2016. How colonization by microbiota in early life shapes the immune system. Science 352:539–544. doi: 10.1126/science.aad9378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bager P, Simonsen J, Nielsen NM, Frisch M. 2012. Cesarean section and offspring's risk of inflammatory bowel disease: a national cohort study. Inflamm Bowel Dis 18:857–862. doi: 10.1002/ibd.21805. [DOI] [PubMed] [Google Scholar]
  • 53.Dotan I, Alper A, Rachmilewitz D, Israeli E, Odes S, Chermesh I, Naftali T, Fraser G, Shitrit AB, Peles V, Reif S. 2013. Maternal inflammatory bowel disease has short and long-term effects on the health of their offspring: a multicenter study in Israel. J Crohns Colitis 7:542–550. doi: 10.1016/j.crohns.2012.08.012. [DOI] [PubMed] [Google Scholar]
  • 54.Örtqvist AK, Lundholm C, Halfvarson J, Ludvigsson JF, Almqvist C. 2019. Fetal and early life antibiotics exposure and very early onset inflammatory bowel disease: a population-based study. Gut 68:218–225. doi: 10.1136/gutjnl-2017-314352. [DOI] [PubMed] [Google Scholar]
  • 55.Romano-Keeler J, Weitkamp JH. 2015. Maternal influences on fetal microbial colonization and immune development. Pediatr Res 77:189–195. doi: 10.1038/pr.2014.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gray LE, O'Hely M, Ranganathan S, Sly PD, Vuillermin P. 2017. The maternal diet, gut bacteria, and bacterial metabolites during pregnancy influence offspring asthma. Front Immunol 8:365. doi: 10.3389/fimmu.2017.00365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, Bjorksten B, Engstrand L, Andersson AF. 2014. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 63:559–566. doi: 10.1136/gutjnl-2012-303249. [DOI] [PubMed] [Google Scholar]
  • 58.Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, Glickman JN, Siebert R, Baron RM, Kasper DL, Blumberg RS. 2012. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336:489–493. doi: 10.1126/science.1219328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Torres J, Hu J, Seki A, Eisele C, Nair N, Huang R, Tarassishin L, Jharap B, Cote-Daigneault J, Mao Q, Mogno I, Britton GJ, Uzzan M, Chen CL, Kornbluth A, George J, Legnani P, Maser E, Loudon H, Stone J, Dubinsky M, Faith JJ, Clemente JC, Mehandru S, Colombel JF, Peter I. 2020. Infants born to mothers with IBD present with altered gut microbiome that transfers abnormalities of the adaptive immune system to germ-free mice. Gut 69:42–51. doi: 10.1136/gutjnl-2018-317855. [DOI] [PubMed] [Google Scholar]
  • 60.Langhorst J, Elsenbruch S, Koelzer J, Rueffer A, Michalsen A, Dobos GJ. 2008. Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol 103:162–169. doi: 10.1111/j.1572-0241.2007.01556.x. [DOI] [PubMed] [Google Scholar]
  • 61.Schoepfer AM, Beglinger C, Straumann A, Trummler M, Vavricka SR, Bruegger LE, Seibold F. 2010. Fecal calprotectin correlates more closely with the Simple Endoscopic Score for Crohn's disease (SES-CD) than CRP, blood leukocytes, and the CDAI. Am J Gastroenterol 105:162–169. doi: 10.1038/ajg.2009.545. [DOI] [PubMed] [Google Scholar]
  • 62.Sipponen T, Savilahti E, Kolho KL, Nuutinen H, Turunen U, Farkkila M. 2008. Crohn's disease activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn's disease activity index and endoscopic findings. Inflamm Bowel Dis 14:40–46. doi: 10.1002/ibd.20312. [DOI] [PubMed] [Google Scholar]
  • 63.Sauter B, Beglinger C, Girardin M, Macpherson A, Michetti P, Schoepfer A, Seibold F, Vavricka SR, Rogler G. 2014. Monitoring disease activity and progression in Crohn's disease. A Swiss perspective on the IBD ahead 'optimised monitoring' recommendations. Digestion 89:299–309. doi: 10.1159/000360283. [DOI] [PubMed] [Google Scholar]
  • 64.Schoepfer AM, Vavricka S, Zahnd-Straumann N, Straumann A, Beglinger C. 2012. Monitoring inflammatory bowel disease activity: clinical activity is judged to be more relevant than endoscopic severity or biomarkers. J Crohns Colitis 6:412–418. doi: 10.1016/j.crohns.2011.09.008. [DOI] [PubMed] [Google Scholar]
  • 65.Derikx JP, Luyer MD, Heineman E, Buurman WA. 2010. Non-invasive markers of gut wall integrity in health and disease. World J Gastroenterol 16:5272–5279. doi: 10.3748/wjg.v16.i42.5272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kim ES, Tarassishin L, Eisele C, Barre A, Nair N, Rendon A, Hawkins K, Debebe A, White S, Thjomoe A, Mork E, Bento-Miranda M, Panchal H, Agrawal M, Patel A, Chen CL, Kornbluth A, George J, Legnani P, Maser E, Loudon H, Mella MT, Stone J, Dubinsky M, Sabino J, Torres J, Colombel JF, Peter I, Hu J, Mount Sinai Road to Prevention Study Group. 2021. Longitudinal changes in fecal calprotectin levels among pregnant women with and without inflammatory bowel disease and their babies. Gastroenterology 160:1118–1130.e3. doi: 10.1053/j.gastro.2020.11.050. [DOI] [PubMed] [Google Scholar]
  • 67.Lundgren SN, Madan JC, Emond JA, Morrison HG, Christensen BC, Karagas MR, Hoen AG. 2018. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome 6:109. doi: 10.1186/s40168-018-0490-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Selma-Royo M, Garcia-Mantrana I, Calatayud M, Parra-Llorca A, Martinez-Costa C, Collado MC. 2021. Maternal diet during pregnancy and intestinal markers are associated with early gut microbiota. Eur J Nutr 60:1429–1442. doi: 10.1007/s00394-020-02337-7. [DOI] [PubMed] [Google Scholar]
  • 69.Garcia-Mantrana I, Selma-Royo M, Gonzalez S, Parra-Llorca A, Martinez-Costa C, Collado MC. 2020. Distinct maternal microbiota clusters are associated with diet during pregnancy: impact on neonatal microbiota and infant growth during the first 18 months of life. Gut Microbes 11:962–978. doi: 10.1080/19490976.2020.1730294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Myklebust-Hansen T, Aamodt G, Haugen M, Brantsæter AL, Vatn MH, Bengtson M-B. 2017. Dietary patterns in women with inflammatory bowel disease and risk of adverse pregnancy outcomes: results from the Norwegian Mother and Child Cohort Study (MoBa). Inflamm Bowel Dis 24:12–24. doi: 10.1093/ibd/izx006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Miller T, Suskind DL. 2018. Exclusive enteral nutrition in pediatric inflammatory bowel disease. Curr Opin Pediatr 30:671–676. doi: 10.1097/MOP.0000000000000660. [DOI] [PubMed] [Google Scholar]
  • 72.Heuschkel RB, Menache CC, Megerian JT, Baird AE. 2000. Enteral nutrition and corticosteroids in the treatment of acute Crohn's disease in children. J Pediatr Gastroenterol Nutr 31:8–15. doi: 10.1097/00005176-200007000-00005. [DOI] [PubMed] [Google Scholar]
  • 73.Borrelli O, Cordischi L, Cirulli M, Paganelli M, Labalestra V, Uccini S, Russo PM, Cucchiara S. 2006. Polymeric diet alone versus corticosteroids in the treatment of active pediatric Crohn's disease: a randomized controlled open-label trial. Clin Gastroenterol Hepatol 4:744–753. doi: 10.1016/j.cgh.2006.03.010. [DOI] [PubMed] [Google Scholar]
  • 74.Berni Canani R, Terrin G, Borrelli O, Romano MT, Manguso F, Coruzzo A, D’Armiento F, Romeo EF, Cucchiara S. 2006. Short- and long-term therapeutic efficacy of nutritional therapy and corticosteroids in paediatric Crohn's disease. Dig Liver Dis 38:381–387. doi: 10.1016/j.dld.2005.10.005. [DOI] [PubMed] [Google Scholar]
  • 75.Swaminath A, Feathers A, Ananthakrishnan AN, Falzon L, Li Ferry S. 2017. Systematic review with meta-analysis: enteral nutrition therapy for the induction of remission in paediatric Crohn's disease. Aliment Pharmacol Ther 46:645–656. doi: 10.1111/apt.14253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Grover Z, Burgess C, Muir R, Reilly C, Lewindon PJ. 2016. Early mucosal healing with exclusive enteral nutrition is associated with improved outcomes in newly diagnosed children with luminal Crohn's disease. J Crohns Colitis 10:1159–1164. doi: 10.1093/ecco-jcc/jjw075. [DOI] [PubMed] [Google Scholar]
  • 77.Yu Y, Chen KC, Chen J. 2019. Exclusive enteral nutrition versus corticosteroids for treatment of pediatric Crohn's disease: a meta-analysis. World J Pediatr 15:26–36. doi: 10.1007/s12519-018-0204-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Narula N, Dhillon A, Zhang D, Sherlock ME, Tondeur M, Zachos M. 2018. Enteral nutritional therapy for induction of remission in Crohn's disease. Cochrane Database Syst Rev 4:CD000542. doi: 10.1002/14651858.CD000542.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Quince C, Ijaz UZ, Loman N, Eren AM, Saulnier D, Russell J, Haig SJ, Calus ST, Quick J, Barclay A, Bertz M, Blaut M, Hansen R, McGrogan P, Russell RK, Edwards CA, Gerasimidis K. 2015. Extensive modulation of the fecal metagenome in children with Crohn's disease during exclusive enteral nutrition. Am J Gastroenterol 110:1718–1729. doi: 10.1038/ajg.2015.357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Diederen K, Li JV, Donachie GE, de Meij TG, de Waart DR, Hakvoort TBM, Kindermann A, Wagner J, Auyeung V, Te Velde AA, Heinsbroek SEM, Benninga MA, Kinross J, Walker AW, de Jonge WJ, Seppen J. 2020. Exclusive enteral nutrition mediates gut microbial and metabolic changes that are associated with remission in children with Crohn's disease. Sci Rep 10:18879. doi: 10.1038/s41598-020-75306-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wahlstrom A, Sayin SI, Marschall HU, Backhed F. 2016. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 24:41–50. doi: 10.1016/j.cmet.2016.05.005. [DOI] [PubMed] [Google Scholar]
  • 82.Ridlon JM, Kang DJ, Hylemon PB. 2006. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47:241–259. doi: 10.1194/jlr.R500013-JLR200. [DOI] [PubMed] [Google Scholar]
  • 83.Wells JE, Berr F, Thomas LA, Dowling RH, Hylemon PB. 2000. Isolation and characterization of cholic acid 7alpha-dehydroxylating fecal bacteria from cholesterol gallstone patients. J Hepatol 32:4–10. doi: 10.1016/S0168-8278(00)80183-X. [DOI] [PubMed] [Google Scholar]
  • 84.Wells JE, Williams KB, Whitehead TR, Heuman DM, Hylemon PB. 2003. Development and application of a polymerase chain reaction assay for the detection and enumeration of bile acid 7alpha-dehydroxylating bacteria in human feces. Clin Chim Acta 331:127–134. doi: 10.1016/S0009-8981(03)00115-3. [DOI] [PubMed] [Google Scholar]
  • 85.Guzior DV, Quinn RA. 2021. Review: microbial transformations of human bile acids. Microbiome 9:140. doi: 10.1186/s40168-021-01101-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Connors J, Dunn KA, Allott J, Bandsma R, Rashid M, Otley AR, Bielawski JP, Van Limbergen J. 2020. The relationship between fecal bile acids and microbiome community structure in pediatric Crohn's disease. ISME J 14:702–713. doi: 10.1038/s41396-019-0560-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Obih C, Wahbeh G, Lee D, Braly K, Giefer M, Shaffer ML, Nielson H, Suskind DL. 2016. Specific carbohydrate diet for pediatric inflammatory bowel disease in clinical practice within an academic IBD center. Nutrition 32:418–425. doi: 10.1016/j.nut.2015.08.025. [DOI] [PubMed] [Google Scholar]
  • 88.Burgis JC, Nguyen K, Park KT, Cox K. 2016. Response to strict and liberalized specific carbohydrate diet in pediatric Crohn's disease. World J Gastroenterol 22:2111–2117. doi: 10.3748/wjg.v22.i6.2111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Suskind DL, Cohen SA, Brittnacher MJ, Wahbeh G, Lee D, Shaffer ML, Braly K, Hayden HS, Klein J, Gold B, Giefer M, Stallworth A, Miller SI. 2018. Clinical and fecal microbial changes with diet therapy in active inflammatory bowel disease. J Clin Gastroenterol 52:155–163. doi: 10.1097/MCG.0000000000000772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, Casero D, Courtney H, Gonzalez A, Graeber TG, Hall AB, Lake K, Landers CJ, Mallick H, Plichta DR, Prasad M, Rahnavard G, Sauk J, Shungin D, Vazquez-Baeza Y, White RA, 3rd, Braun J, Denson LA, Jansson JK, Knight R, Kugathasan S, McGovern DPB, Petrosino JF, Stappenbeck TS, Winter HS, Clish CB, Franzosa EA, Vlamakis H, Xavier RJ, Huttenhower C, IMDMDB Investigators. 2019. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569:655–662. doi: 10.1038/s41586-019-1237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Geerling BJ, Badart-Smook A, Stockbrugger RW, Brummer RJ. 1998. Comprehensive nutritional status in patients with long-standing Crohn disease currently in remission. Am J Clin Nutr 67:919–926. doi: 10.1093/ajcn/67.5.919. [DOI] [PubMed] [Google Scholar]
  • 92.Filippi J, Al-Jaouni R, Wiroth JB, Hebuterne X, Schneider SM. 2006. Nutritional deficiencies in patients with Crohn's disease in remission. Inflamm Bowel Dis 12:185–191. doi: 10.1097/01.MIB.0000206541.15963.c3. [DOI] [PubMed] [Google Scholar]
  • 93.Massironi S, Rossi RE, Cavalcoli FA, Della Valle S, Fraquelli M, Conte D. 2013. Nutritional deficiencies in inflammatory bowel disease: therapeutic approaches. Clin Nutr 32:904–910. doi: 10.1016/j.clnu.2013.03.020. [DOI] [PubMed] [Google Scholar]
  • 94.Seidman E, LeLeiko N, Ament M, Berman W, Caplan D, Evans J, Kocoshis S, Lake A, Motil K, Sutphen J. 1991. Nutritional issues in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 12:424–438. doi: 10.1097/00005176-199105000-00004. [DOI] [PubMed] [Google Scholar]
  • 95.Jowett SL, Seal CJ, Phillips E, Gregory W, Barton JR, Welfare MR. 2004. Dietary beliefs of people with ulcerative colitis and their effect on relapse and nutrient intake. Clin Nutr 23:161–170. doi: 10.1016/S0261-5614(03)00132-8. [DOI] [PubMed] [Google Scholar]
  • 96.Braly K, Williamson N, Shaffer ML, Lee D, Wahbeh G, Klein J, Giefer M, Suskind DL. 2017. Nutritional adequacy of the specific carbohydrate diet in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 65:533–538. doi: 10.1097/MPG.0000000000001613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Fritsch J, Garces L, Quintero MA, Pignac-Kobinger J, Santander AM, Fernandez I, Ban YJ, Kwon D, Phillips MC, Knight K, Mao Q, Santaolalla R, Chen XS, Maruthamuthu M, Solis N, Damas OM, Kerman DH, Deshpande AR, Lewis JE, Chen C, Abreu MT. 2020. Low-fat, high-fiber diet reduces markers of inflammation and dysbiosis and improves quality of life in patients with ulcerative colitis. Clin Gastroenterol Hepatol 19:1189–1199.e30. doi: 10.1016/j.cgh.2020.05.026. [DOI] [PubMed] [Google Scholar]
  • 98.Cox SR, Lindsay JO, Fromentin S, Stagg AJ, McCarthy NE, Galleron N, Ibraim SB, Roume H, Levenez F, Pons N, Maziers N, Lomer MC, Ehrlich SD, Irving PM, Whelan K. 2020. Effects of low FODMAP diet on symptoms, fecal microbiome, and markers of inflammation in patients with quiescent inflammatory bowel disease in a Randomized trial. Gastroenterology 158:176–188.e7. doi: 10.1053/j.gastro.2019.09.024. [DOI] [PubMed] [Google Scholar]
  • 99.Grammatikopoulou MG, Goulis DG, Gkiouras K, Nigdelis MP, Papageorgiou ST, Papamitsou T, Forbes A, Bogdanos DP. 2020. Low FODMAP diet for functional gastrointestinal symptoms in quiescent inflammatory bowel disease: a systematic review of randomized controlled trials. Nutrients 12:3648. doi: 10.3390/nu12123648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Bodini G, Zanella C, Crespi M, Lo Pumo S, Demarzo MG, Savarino E, Savarino V, Giannini EG. 2019. A randomized, 6-wk trial of a low FODMAP diet in patients with inflammatory bowel disease. Nutrition 67–68:110542. doi: 10.1016/j.nut.2019.06.023. [DOI] [PubMed] [Google Scholar]
  • 101.Gibson PR. 2017. Use of the low-FODMAP diet in inflammatory bowel disease. J Gastroenterol Hepatol 32(Suppl 1):40–42. doi: 10.1111/jgh.13695. [DOI] [PubMed] [Google Scholar]
  • 102.Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van Immerseel F, Verbeke K, Ferrante M, Verhaegen J, Rutgeerts P, Vermeire S. 2014. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 63:1275–1283. doi: 10.1136/gutjnl-2013-304833. [DOI] [PubMed] [Google Scholar]
  • 103.Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, Vandamme P, Vermeire S. 2011. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut 60:631–637. doi: 10.1136/gut.2010.223263. [DOI] [PubMed] [Google Scholar]
  • 104.Pascal V, Pozuelo M, Borruel N, Casellas F, Campos D, Santiago A, Martinez X, Varela E, Sarrabayrouse G, Machiels K, Vermeire S, Sokol H, Guarner F, Manichanh C. 2017. A microbial signature for Crohn's disease. Gut 66:813–822. doi: 10.1136/gutjnl-2016-313235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kumari R, Ahuja V, Paul J. 2013. Fluctuations in butyrate-producing bacteria in ulcerative colitis patients of North India. World J Gastroenterol 19:3404–3414. doi: 10.3748/wjg.v19.i22.3404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M, Morgan XC, Kostic AD, Luo C, Gonzalez A, McDonald D, Haberman Y, Walters T, Baker S, Rosh J, Stephens M, Heyman M, Markowitz J, Baldassano R, Griffiths A, Sylvester F, Mack D, Kim S, Crandall W, Hyams J, Huttenhower C, Knight R, Xavier RJ. 2014. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 15:382–392. doi: 10.1016/j.chom.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lewis JD, Chen EZ, Baldassano RN, Otley AR, Griffiths AM, Lee D, Bittinger K, Bailey A, Friedman ES, Hoffmann C, Albenberg L, Sinha R, Compher C, Gilroy E, Nessel L, Grant A, Chehoud C, Li H, Wu GD, Bushman FD. 2015. Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn's disease. Cell Host Microbe 18:489–500. doi: 10.1016/j.chom.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. 2007. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA 104:13780–13785. doi: 10.1073/pnas.0706625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Baumgart M, Dogan B, Rishniw M, Weitzman G, Bosworth B, Yantiss R, Orsi RH, Wiedmann M, McDonough P, Kim SG, Berg D, Schukken Y, Scherl E, Simpson KW. 2007. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn's disease involving the ileum. ISME J 1:403–418. doi: 10.1038/ismej.2007.52. [DOI] [PubMed] [Google Scholar]
  • 110.Lepage P, Hasler R, Spehlmann ME, Rehman A, Zvirbliene A, Begun A, Ott S, Kupcinskas L, Dore J, Raedler A, Schreiber S. 2011. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology 141:227–236. doi: 10.1053/j.gastro.2011.04.011. [DOI] [PubMed] [Google Scholar]
  • 111.Haberman Y, Tickle TL, Dexheimer PJ, Kim MO, Tang D, Karns R, Baldassano RN, Noe JD, Rosh J, Markowitz J, Heyman MB, Griffiths AM, Crandall WV, Mack DR, Baker SS, Huttenhower C, Keljo DJ, Hyams JS, Kugathasan S, Walters TD, Aronow B, Xavier RJ, Gevers D, Denson LA. 2014. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J Clin Invest 124:3617–3633. doi: 10.1172/JCI75436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Halfvarson J, Brislawn CJ, Lamendella R, Vazquez-Baeza Y, Walters WA, Bramer LM, D'Amato M, Bonfiglio F, McDonald D, Gonzalez A, McClure EE, Dunklebarger MF, Knight R, Jansson JK. 2017. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat Microbiol 2:17004. doi: 10.1038/nmicrobiol.2017.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Mondot S, Kang S, Furet JP, Aguirre de Carcer D, McSweeney C, Morrison M, Marteau P, Dore J, Leclerc M. 2011. Highlighting new phylogenetic specificities of Crohn's disease microbiota. Inflamm Bowel Dis 17:185–192. doi: 10.1002/ibd.21436. [DOI] [PubMed] [Google Scholar]
  • 114.Pickert G, Wirtz S, Matzner J, Ashfaq-Khan M, Heck R, Rosigkeit S, Thies D, Surabattula R, Ehmann D, Wehkamp J, Aslam M, He G, Weigert A, Foerster F, Klotz L, Frick JS, Becker C, Bockamp E, Schuppan D. 2020. Wheat consumption aggravates colitis in mice via amylase trypsin inhibitor-mediated dysbiosis. Gastroenterology 159:257–272.e17. doi: 10.1053/j.gastro.2020.03.064. [DOI] [PubMed] [Google Scholar]
  • 115.Zevallos VF, Raker V, Tenzer S, Jimenez-Calvente C, Ashfaq-Khan M, Russel N, Pickert G, Schild H, Steinbrink K, Schuppan D. 2017. Nutritional wheat amylase-trypsin inhibitors promote intestinal inflammation via activation of myeloid cells. Gastroenterology 152:1100–1113.e12. doi: 10.1053/j.gastro.2016.12.006. [DOI] [PubMed] [Google Scholar]
  • 116.Caminero A, McCarville JL, Zevallos VF, Pigrau M, Yu XB, Jury J, Galipeau HJ, Clarizio AV, Casqueiro J, Murray JA, Collins SM, Alaedini A, Bercik P, Schuppan D, Verdu EF. 2019. Lactobacilli degrade wheat amylase trypsin inhibitors to reduce intestinal dysfunction induced by immunogenic wheat proteins. Gastroenterology 156:2266–2280. doi: 10.1053/j.gastro.2019.02.028. [DOI] [PubMed] [Google Scholar]
  • 117.Green N, Miller T, Suskind D, Lee D. 2019. A review of dietary therapy for IBD and a vision for the future. Nutrients 11:947. doi: 10.3390/nu11050947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.de Castro MM, Corona LP, Pascoal LB, Miyamoto JE, Ignacio-Souza LM, de Lourdes Setsuko Ayrizono M, Torsoni MA, Torsoni AS, Leal RF, Milanski M. 2020. Dietary patterns associated to clinical aspects in Crohn's disease patients. Sci Rep 10:7033. doi: 10.1038/s41598-020-64024-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Deehan EC, Yang C, Perez-Munoz ME, Nguyen NK, Cheng CC, Triador L, Zhang Z, Bakal JA, Walter J. 2020. Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host Microbe 27:389–404.e6. doi: 10.1016/j.chom.2020.01.006. [DOI] [PubMed] [Google Scholar]
  • 120.Barbara O, Bucci Vanni CC, Rene M, Margaret M, Effie O, Camilla M, David C, Ward Doyle V, Randall P, Christine F, Shakti B, McCormick Beth A, Maldonado-Contreras A. 2021. Dietary manipulation of the gut microbiome in inflammatory bowel disease patients: proof of concept. medRXiv. 10.1101/2021.10.07.21250296. [DOI] [Google Scholar]
  • 121.Alexander M, Turnbaugh PJ. 2020. Deconstructing mechanisms of diet-microbiome-immune interactions. Immunity 53:264–276. doi: 10.1016/j.immuni.2020.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zeng MY, Inohara N, Nunez G. 2017. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol 10:18–26. doi: 10.1038/mi.2016.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Alhmoud T, Kumar A, Lo C-C, Al-Sadi R, Clegg S, Alomari I, Zmeili T, Gleasne CD, Mcmurry K, Dichosa AEK, Vuyisich M, Chain PSG, Mishra S, Ma T. 2019. Investigating intestinal permeability and gut microbiota roles in acute coronary syndrome patients. Hum Microb J 13:100059. doi: 10.1016/j.humic.2019.100059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Pedersen C, Ijaz UZ, Gallagher E, Horton F, Ellis RJ, Jaiyeola E, Duparc T, Russell-Jones D, Hinton P, Cani PD, La Ragione RM, Robertson MD. 2018. Fecal Enterobacteriales enrichment is associated with increased in vivo intestinal permeability in humans. Physiol Rep 6:e13649. doi: 10.14814/phy2.13649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Baldelli V, Scaldaferri F, Putignani L, Del Chierico F. 2021. The role of Enterobacteriaceae in gut microbiota dysbiosis in inflammatory bowel diseases. Microorganisms 9:697. doi: 10.3390/microorganisms9040697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Thoo L, Noti M, Krebs P. 2019. Keep calm: the intestinal barrier at the interface of peace and war. Cell Death Dis 10:849. doi: 10.1038/s41419-019-2086-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Odenwald MA, Turner JR. 2013. Intestinal permeability defects: is it time to treat? Clin Gastroenterol Hepatol 11:1075–1083. doi: 10.1016/j.cgh.2013.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sommer K, Wiendl M, Muller TM, Heidbreder K, Voskens C, Neurath MF, Zundler S. 2021. Intestinal mucosal wound healing and barrier integrity in IBD-crosstalk and trafficking of cellular players. Front Med (Lausanne) 8:643973. doi: 10.3389/fmed.2021.643973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Artis D. 2008. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol 8:411–420. doi: 10.1038/nri2316. [DOI] [PubMed] [Google Scholar]
  • 130.May GR, Sutherland LR, Meddings JB. 1993. Is small intestinal permeability really increased in relatives of patients with Crohn's disease? Gastroenterology 104:1627–1632. doi: 10.1016/0016-5085(93)90638-s. [DOI] [PubMed] [Google Scholar]
  • 131.Hollander D, Vadheim CM, Brettholz E, Petersen GM, Delahunty T, Rotter JI. 1986. Increased intestinal permeability in patients with Crohn's disease and their relatives. A possible etiologic factor. Ann Intern Med 105:883–885. doi: 10.7326/0003-4819-105-6-883. [DOI] [PubMed] [Google Scholar]
  • 132.Irvine EJ, Marshall JK. 2000. Increased intestinal permeability precedes the onset of Crohn's disease in a subject with familial risk. Gastroenterology 119:1740–1744. doi: 10.1053/gast.2000.20231. [DOI] [PubMed] [Google Scholar]
  • 133.Turpin W, Lee S-H, Raygoza Garay JA, Madsen KL, Meddings JB, Bedrani L, Power N, Espin-Garcia O, Xu W, Smith MI, Griffiths AM, Moayyedi P, Turner D, Seidman EG, Steinhart AH, Marshall JK, Jacobson K, Mack D, Huynh H, Bernstein CN, Paterson AD, Croitoru K, Crohn's and Colitis Canada Genetic Environmental Microbial Project Research Consortium, CCC GEM Project recruitment site directors Maria Abreu and Kenneth Croitoru. 2020. Increased intestinal permeability is associated with later development of Crohn's disease. Gastroenterology 159:2092–2100.e5. doi: 10.1053/j.gastro.2020.08.005. [DOI] [PubMed] [Google Scholar]
  • 134.Galipeau HJ, Caminero A, Turpin W, Bermudez-Brito M, Santiago A, Libertucci J, Constante M, Raygoza Garay JA, Rueda G, Armstrong S, Clarizio A, Smith MI, Surette MG, Bercik P, Croitoru K, Verdu EF, CCC Genetics Environmental, Microbial Project Research Consortium, Croitoru K, Verdu EF. 2021. Novel fecal biomarkers that precede clinical diagnosis of ulcerative colitis. Gastroenterology 160:1532–1545. doi: 10.1053/j.gastro.2020.12.004. [DOI] [PubMed] [Google Scholar]
  • 135.Van Spaendonk H, Ceuleers H, Witters L, Patteet E, Joossens J, Augustyns K, Lambeir AM, De Meester I, De Man JG, De Winter BY. 2017. Regulation of intestinal permeability: the role of proteases. World J Gastroenterol 23:2106–2123. doi: 10.3748/wjg.v23.i12.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Pullan RD, Thomas GA, Rhodes M, Newcombe RG, Williams GT, Allen A, Rhodes J. 1994. Thickness of adherent mucus gel on colonic mucosa in humans and its relevance to colitis. Gut 35:353–359. doi: 10.1136/gut.35.3.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Courth LF, Ostaff MJ, Mailander-Sanchez D, Malek NP, Stange EF, Wehkamp J. 2015. Crohn's disease-derived monocytes fail to induce Paneth cell defensins. Proc Natl Acad Sci USA 112:14000–14005. doi: 10.1073/pnas.1510084112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wehkamp J, Koslowski M, Wang G, Stange EF. 2008. Barrier dysfunction due to distinct defensin deficiencies in small intestinal and colonic Crohn's disease. Mucosal Immunol 1(Suppl 1):S67–S74. doi: 10.1038/mi.2008.48. [DOI] [PubMed] [Google Scholar]
  • 139.Vivinus-Nebot M, Frin-Mathy G, Bzioueche H, Dainese R, Bernard G, Anty R, Filippi J, Saint-Paul MC, Tulic MK, Verhasselt V, Hebuterne X, Piche T. 2014. Functional bowel symptoms in quiescent inflammatory bowel diseases: role of epithelial barrier disruption and low-grade inflammation. Gut 63:744–752. doi: 10.1136/gutjnl-2012-304066. [DOI] [PubMed] [Google Scholar]
  • 140.Chang J, Leong RW, Wasinger VC, Ip M, Yang M, Phan TG. 2017. Impaired intestinal permeability contributes to ongoing bowel symptoms in patients with inflammatory bowel disease and mucosal healing. Gastroenterology 153:723–731.e1. doi: 10.1053/j.gastro.2017.05.056. [DOI] [PubMed] [Google Scholar]
  • 141.Wyatt J, Vogelsang H, Hubl W, Waldhoer T, Lochs H. 1993. Intestinal permeability and the prediction of relapse in Crohn's disease. Lancet 341:1437–1439. doi: 10.1016/0140-6736(93)90882-H. [DOI] [PubMed] [Google Scholar]
  • 142.Arnott ID, Kingstone K, Ghosh S. 2000. Abnormal intestinal permeability predicts relapse in inactive Crohn disease. Scand J Gastroenterol 35:1163–1169. doi: 10.1080/003655200750056637. [DOI] [PubMed] [Google Scholar]
  • 143.D'Inca R, Di Leo V, Corrao G, Martines D, D'Odorico A, Mestriner C, Venturi C, Longo G, Sturniolo GC. 1999. Intestinal permeability test as a predictor of clinical course in Crohn's disease. Am J Gastroenterol 94:2956–2960. doi: 10.1111/j.1572-0241.1999.01444.x. [DOI] [PubMed] [Google Scholar]
  • 144.Donohoe DR, Garge N, Zhang X, Sun W, O'Connell TM, Bunger MK, Bultman SJ. 2011. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab 13:517–526. doi: 10.1016/j.cmet.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Huang X, Oshima T, Tomita T, Fukui H, Miwa H. 2021. Butyrate alleviates cytokine-induced barrier dysfunction by modifying claudin-2 levels. Biology (Basel) 10:205. doi: 10.3390/biology10030205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wang HB, Wang PY, Wang X, Wan YL, Liu YC. 2012. Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein Claudin-1 transcription. Dig Dis Sci 57:3126–3135. doi: 10.1007/s10620-012-2259-4. [DOI] [PubMed] [Google Scholar]
  • 147.Chelakkot C, Ghim J, Ryu SH. 2018. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med 50:1–9. doi: 10.1038/s12276-018-0126-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, Weir TL, Ehrentraut SF, Pickel C, Kuhn KA, Lanis JM, Nguyen V, Taylor CT, Colgan SP. 2015. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17:662–671. doi: 10.1016/j.chom.2015.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Deatherage Kaiser BL, Li J, Sanford JA, Kim YM, Kronewitter SR, Jones MB, Peterson CT, Peterson SN, Frank BC, Purvine SO, Brown JN, Metz TO, Smith RD, Heffron F, Adkins JN. 2013. A multi-omic view of host-pathogen-commensal interplay in Salmonella-mediated intestinal infection. PLoS One 8:e67155. doi: 10.1371/journal.pone.0067155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Schroeder BO, Birchenough GMH, Stahlman M, Arike L, Johansson MEV, Hansson GC, Backhed F. 2018. Bifidobacteria or fiber protects against diet-induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe 23:27–40.e7. doi: 10.1016/j.chom.2017.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck TS, Nunez G, Martens EC. 2016. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167:1339–1353.e21. doi: 10.1016/j.cell.2016.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Derrien M, Vaughan EE, Plugge CM, de Vos WM. 2004. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 54:1469–1476. doi: 10.1099/ijs.0.02873-0. [DOI] [PubMed] [Google Scholar]
  • 153.El Kaoutari A, Armougom F, Gordon JI, Raoult D, Henrissat B. 2013. The abundance and variety of carbohydrate-active enzymes in the human gut microbiota. Nat Rev Microbiol 11:497–504. doi: 10.1038/nrmicro3050. [DOI] [PubMed] [Google Scholar]
  • 154.Xie R, Sun Y, Wu J, Huang S, Jin G, Guo Z, Zhang Y, Liu T, Liu X, Cao X, Wang B, Cao H. 2018. Maternal high fat diet alters gut microbiota of offspring and exacerbates DSS-induced colitis in adulthood. Front Immunol 9:2608. doi: 10.3389/fimmu.2018.02608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Chelakkot C, Ghim J, Rajasekaran N, Choi JS, Kim JH, Jang MH, Shin YK, Suh PG, Ryu SH. 2017. Intestinal epithelial cell-specific deletion of PLD2 alleviates DSS-induced colitis by regulating occludin. Sci Rep 7:1573. doi: 10.1038/s41598-017-01797-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yamamoto-Furusho JK, Mendivil EJ, Mendivil-Rangel EJ, Fonseca-Camarillo G. 2012. Differential expression of occludin in patients with ulcerative colitis and healthy controls. Inflamm Bowel Dis 18:E1999. doi: 10.1002/ibd.22835. [DOI] [PubMed] [Google Scholar]
  • 157.Kucharzik T, Walsh SV, Chen J, Parkos CA, Nusrat A. 2001. Neutrophil transmigration in inflammatory bowel disease is associated with differential expression of epithelial intercellular junction proteins. Am J Pathol 159:2001–2009. doi: 10.1016/S0002-9440(10)63051-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Valenzano MC, DiGuilio K, Mercado J, Teter M, To J, Ferraro B, Mixson B, Manley I, Baker V, Moore BA, Wertheimer J, Mullin JM. 2015. Remodeling of tight junctions and enhancement of barrier integrity of the CACO-2 intestinal epithelial cell layer by micronutrients. PLoS One 10:e0133926. doi: 10.1371/journal.pone.0133926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gruber L, Kisling S, Lichti P, Martin FP, May S, Klingenspor M, Lichtenegger M, Rychlik M, Haller D. 2013. High fat diet accelerates pathogenesis of murine Crohn's disease-like ileitis independently of obesity. PLoS One 8:e71661. doi: 10.1371/journal.pone.0071661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Llewellyn SR, Britton GJ, Contijoch EJ, Vennaro OH, Mortha A, Colombel JF, Grinspan A, Clemente JC, Merad M, Faith JJ. 2018. Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice. Gastroenterology 154:1037–1046.e2. doi: 10.1053/j.gastro.2017.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Roberts CL, Keita AV, Duncan SH, O'Kennedy N, Soderholm JD, Rhodes JM, Campbell BJ. 2010. Translocation of Crohn's disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut 59:1331–1339. doi: 10.1136/gut.2009.195370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, Gewirtz AT. 2015. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 519:92–96. doi: 10.1038/nature14232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, Gewirtz AT. 2016. Corrigendum: dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 536:238. doi: 10.1038/nature18000. [DOI] [PubMed] [Google Scholar]
  • 164.Lock JY, Carlson TL, Wang CM, Chen A, Carrier RL. 2018. Acute exposure to commonly ingested emulsifiers alters intestinal mucus structure and transport properties. Sci Rep 8:10008. doi: 10.1038/s41598-018-27957-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Viennois E, Bretin A, Dube PE, Maue AC, Dauriat CJG, Barnich N, Gewirtz AT, Chassaing B. 2020. Dietary emulsifiers directly impact adherent-invasive E. coli gene expression to drive chronic intestinal inflammation. Cell Rep 33:108229. doi: 10.1016/j.celrep.2020.108229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Panduro M, Benoist C, Mathis D. 2016. Tissue Tregs. Annu Rev Immunol 34:609–633. doi: 10.1146/annurev-immunol-032712-095948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Round JL, Mazmanian SK. 2010. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA 107:12204–12209. doi: 10.1073/pnas.0909122107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Bollrath J, Powrie FM. 2013. Controlling the frontier: regulatory T-cells and intestinal homeostasis. Semin Immunol 25:352–357. doi: 10.1016/j.smim.2013.09.002. [DOI] [PubMed] [Google Scholar]
  • 169.Fantini MC, Becker C, Tubbe I, Nikolaev A, Lehr HA, Galle P, Neurath MF. 2006. Transforming growth factor beta induced FoxP3+ regulatory T cells suppress Th1 mediated experimental colitis. Gut 55:671–680. doi: 10.1136/gut.2005.072801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Boden EK, Snapper SB. 2008. Regulatory T cells in inflammatory bowel disease. Curr Opin Gastroenterol 24:733–741. doi: 10.1097/mog.0b013e328311f26e. [DOI] [PubMed] [Google Scholar]
  • 171.Stein RR, Tanoue T, Szabady RL, Bhattarai SK, Olle B, Norman JM, Suda W, Oshima K, Hattori M, Gerber GK, Sander C, Honda K, Bucci V. 2018. Computer-guided design of optimal microbial consortia for immune system modulation. Elife 7:e30916. doi: 10.7554/eLife.30916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Nagano Y, Itoh K, Honda K. 2012. The induction of Treg cells by gut-indigenous Clostridium. Curr Opin Immunol 24:392–397. doi: 10.1016/j.coi.2012.05.007. [DOI] [PubMed] [Google Scholar]
  • 173.Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, Geva-Zatorsky N, Jupp R, Mathis D, Benoist C, Kasper DL. 2020. Microbial bile acid metabolites modulate gut RORgamma(+) regulatory T cell homeostasis. Nature 577:410–415. doi: 10.1038/s41586-019-1865-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A, Antonopoulos DA, Jabri B, Chang EB. 2012. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 487:104–108. doi: 10.1038/nature11225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Hardison WG. 1978. Hepatic taurine concentration and dietary taurine as regulators of bile acid conjugation with taurine. Gastroenterology 75:71–75. doi: 10.1016/0016-5085(78)93767-8. [DOI] [PubMed] [Google Scholar]
  • 176.Sjovall J. 1959. Dietary glycine and taurine on bile acid conjugation in man; bile acids and steroids 75. Proc Soc Exp Biol Med 100:676–678. doi: 10.3181/00379727-100-24741. [DOI] [PubMed] [Google Scholar]
  • 177.Magee EA, Richardson CJ, Hughes R, Cummings JH. 2000. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr 72:1488–1494. doi: 10.1093/ajcn/72.6.1488. [DOI] [PubMed] [Google Scholar]
  • 178.Sinha SR, Haileselassie Y, Nguyen LP, Tropini C, Wang M, Becker LS, Sim D, Jarr K, Spear ET, Singh G, Namkoong H, Bittinger K, Fischbach MA, Sonnenburg JL, Habtezion A. 2020. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 27:659–670. doi: 10.1016/j.chom.2020.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, Takei H, Muto A, Nittono H, Ridlon JM, White MB, Noble NA, Monteith P, Fuchs M, Thacker LR, Sikaroodi M, Bajaj JS. 2013. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 58:949–955. doi: 10.1016/j.jhep.2013.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Theriot CM, Bowman AA, Young VB. 2016. Antibiotic-induced alterations of the gut microbiota alter secondary bile acid production and allow for Clostridium difficile spore germination and outgrowth in the large intestine. mSphere 1:e00045-15. doi: 10.1128/mSphere.00045-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Yoneno K, Hisamatsu T, Shimamura K, Kamada N, Ichikawa R, Kitazume MT, Mori M, Uo M, Namikawa Y, Matsuoka K, Sato T, Koganei K, Sugita A, Kanai T, Hibi T. 2013. TGR5 signalling inhibits the production of pro-inflammatory cytokines by in vitro differentiated inflammatory and intestinal macrophages in Crohn's disease. Immunology 139:19–29. doi: 10.1111/imm.12045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Nakajima A, Kaga N, Nakanishi Y, Ohno H, Miyamoto J, Kimura I, Hori S, Sasaki T, Hiramatsu K, Okumura K, Miyake S, Habu S, Watanabe S. 2017. Maternal high fiber diet during pregnancy and lactation influences regulatory T cell differentiation in offspring in mice. J Immunol 199:3516–3524. doi: 10.4049/jimmunol.1700248. [DOI] [PubMed] [Google Scholar]
  • 183.Wang H, Shi P, Zuo L, Dong J, Zhao J, Liu Q, Zhu W. 2016. Dietary non-digestible polysaccharides ameliorate intestinal epithelial barrier dysfunction in IL-10 knockout mice. J Crohns Colitis 10:1076–1086. doi: 10.1093/ecco-jcc/jjw065. [DOI] [PubMed] [Google Scholar]
  • 184.Yang H, Youm YH, Vandanmagsar B, Rood J, Kumar KG, Butler AA, Dixit VD. 2009. Obesity accelerates thymic aging. Blood 114:3803–3812. doi: 10.1182/blood-2009-03-213595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Zhang M, Zhou Q, Dorfman RG, Huang X, Fan T, Zhang H, Zhang J, Yu C. 2016. Butyrate inhibits interleukin-17 and generates Tregs to ameliorate colorectal colitis in rats. BMC Gastroenterol 16:84. doi: 10.1186/s12876-016-0500-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H. 2013. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504:446–450. doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
  • 187.Zhai S, Qin S, Li L, Zhu L, Zou Z, Wang L. 2019. Dietary butyrate suppresses inflammation through modulating gut microbiota in high-fat diet-fed mice. FEMS Microbiol Lett 366:fnz153. doi: 10.1093/femsle/fnz153. [DOI] [PubMed] [Google Scholar]
  • 188.Schulthess J, Pandey S, Capitani M, Rue-Albrecht KC, Arnold I, Franchini F, Chomka A, Ilott NE, Johnston DGW, Pires E, McCullagh J, Sansom SN, Arancibia-Carcamo CV, Uhlig HH, Powrie F. 2019. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 50:432–445.e7. doi: 10.1016/j.immuni.2018.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. 2013. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 145:396–406.e1-10. doi: 10.1053/j.gastro.2013.04.056. [DOI] [PubMed] [Google Scholar]
  • 190.Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, Kim CH. 2015. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol 8:80–93. doi: 10.1038/mi.2014.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Afzali A, Cross RK. 2016. Racial and ethnic minorities with inflammatory bowel disease in the United States: a systematic review of disease characteristics and differences. Inflamm Bowel Dis 22:2023–2040. doi: 10.1097/MIB.0000000000000835. [DOI] [PubMed] [Google Scholar]
  • 192.Appleyard CB, Hernandez G, Rios-Bedoya CF. 2004. Basic epidemiology of inflammatory bowel disease in Puerto Rico. Inflamm Bowel Dis 10:106–111. doi: 10.1097/00054725-200403000-00007. [DOI] [PubMed] [Google Scholar]
  • 193.U.S. Census Bureau. 2013. Asians fastest-growing race or ethnic group in 2012, Census Bureau reports. Release no. CB13-112. U.S. Census Bureau. [Google Scholar]
  • 194.U.S. Census Bureau. 2020. Improved race and ethnicity measures reveal U.S. population is much more multiracial. https://www.census.gov/library/stories/2021/08/improved-race-ethnicity-measures-reveal-united-states-population-much-more-multiracial.html.
  • 195.Damas OM, Jahann DA, Reznik R, McCauley JL, Tamariz L, Deshpande AR, Abreu MT, Sussman DA. 2013. Phenotypic manifestations of inflammatory bowel disease differ between Hispanics and non-Hispanic whites: results of a large cohort study. Am J Gastroenterol 108:231–239. doi: 10.1038/ajg.2012.393. [DOI] [PubMed] [Google Scholar]
  • 196.Avalos DJ, Mendoza-Ladd A, Zuckerman MJ, Bashashati M, Alvarado A, Dwivedi A, Damas OM. 2018. Hispanic Americans and non-Hispanic white Americans have a similar inflammatory bowel disease phenotype: a systematic review with meta-analysis. Dig Dis Sci 63:1558–1571. doi: 10.1007/s10620-018-5022-7. [DOI] [PubMed] [Google Scholar]
  • 197.Damas OM, Estes D, Avalos D, Quintero MA, Morillo D, Caraballo F, Lopez J, Deshpande AR, Kerman D, McCauley JL, Palacio A, Abreu MT, Schwartz SJ. 2018. Hispanics coming to the US adopt US cultural behaviors and eat less healthy: implications for development of inflammatory bowel disease. Dig Dis Sci 63:3058–3066. doi: 10.1007/s10620-018-5185-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Jangi S, Ruan A, Korzenik J, de Silva P. 2020. South Asian patients with inflammatory bowel disease in the United States demonstrate more fistulizing and perianal Crohn phenotype. Inflamm Bowel Dis 26:1933–1942. doi: 10.1093/ibd/izaa029. [DOI] [PubMed] [Google Scholar]
  • 199.Ahuja V, Tandon RK. 2010. Inflammatory bowel disease in the Asia-Pacific area: a comparison with developed countries and regional differences. J Dig Dis 11:134–147. doi: 10.1111/j.1751-2980.2010.00429.x. [DOI] [PubMed] [Google Scholar]
  • 200.Vangay P, Johnson AJ, Ward TL, Al-Ghalith GA, Shields-Cutler RR, Hillmann BM, Lucas SK, Beura LK, Thompson EA, Till LM, Batres R, Paw B, Pergament SL, Saenyakul P, Xiong M, Kim AD, Kim G, Masopust D, Martens EC, Angkurawaranon C, McGready R, Kashyap PC, Culhane-Pera KA, Knights D. 2018. US immigration westernizes the human gut microbiome. Cell 175:962–972.e10. doi: 10.1016/j.cell.2018.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Aswani-Omprakash T, Sharma V, Bishu S, Balasubramaniam M, Bhatia S, Nandi N, Shah ND, Deepak P, Sebastian S. 2021. Addressing unmet needs from a new frontier of IBD: the South Asian IBD Alliance. Lancet Gastroenterol Hepatol 6:884–885. doi: 10.1016/S2468-1253(21)00336-8. [DOI] [PubMed] [Google Scholar]
  • 202.Kirby JB, Taliaferro G, Zuvekas SH. 2006. Explaining racial and ethnic disparities in health care. Med Care 44:I64–72. doi: 10.1097/01.mlr.0000208195.83749.c3. [DOI] [PubMed] [Google Scholar]
  • 203.Chen J, Vargas-Bustamante A, Mortensen K, Ortega AN. 2016. Racial and ethnic disparities in health care access and utilization under the Affordable Care Act. Med Care 54:140–146. doi: 10.1097/MLR.0000000000000467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Hoffman KM, Trawalter S, Axt JR, Oliver MN. 2016. Racial bias in pain assessment and treatment recommendations, and false beliefs about biological differences between blacks and whites. Proc Natl Acad Sci USA 113:4296–4301. doi: 10.1073/pnas.1516047113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Vincent D, McEwen MM, Hepworth JT, Stump CS. 2014. The effects of a community-based, culturally tailored diabetes prevention intervention for high-risk adults of Mexican descent. Diabetes Educ 40:202–213. doi: 10.1177/0145721714521020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Rosal MC, Ockene IS, Restrepo A, White MJ, Borg A, Olendzki B, Scavron J, Candib L, Welch G, Reed G. 2011. Randomized trial of a literacy-sensitive, culturally tailored diabetes self-management intervention for low-income Latinos: Latinos en Control. Diabetes Care 34:838–844. doi: 10.2337/dc10-1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kim H, Song HJ, Han HR, Kim KB, Kim MT. 2013. Translation and validation of the Dietary Approaches to Stop Hypertension for Koreans intervention: culturally tailored dietary guidelines for Korean Americans with high blood pressure. J Cardiovasc Nurs 28:514–523. doi: 10.1097/JCN.0b013e318262c0c1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Risica PM, Gans KM, Kumanyika S, Kirtania U, Lasater TM. 2013. SisterTalk: final results of a culturally tailored cable television delivered weight control program for Black women. Int J Behav Nutr Phys Act 10:141. doi: 10.1186/1479-5868-10-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Scarinci IC, Moore A, Wynn-Wallace T, Cherrington A, Fouad M, Li Y. 2014. A community-based, culturally relevant intervention to promote healthy eating and physical activity among middle-aged African American women in rural Alabama: findings from a group randomized controlled trial. Prev Med 69:13–20. doi: 10.1016/j.ypmed.2014.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Kaiser L, Martinez J, Horowitz M, Lamp C, Johns M, Espinoza D, Byrnes M, Gomez MM, Aguilera A, de la Torre A. 2015. Adaptation of a culturally relevant nutrition and physical activity program for low-income, Mexican-origin parents with young children. Prev Chronic Dis 12:E72. doi: 10.5888/pcd12.140591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Flores-Luevano S, Pacheco M, Shokar GS, Dwivedi AK, Shokar NK. 2020. Impact of a culturally tailored diabetes education and empowerment program in a Mexican American population along the US/Mexico border: a pragmatic study. J Clin Med Res 12:517–529. doi: 10.14740/jocmr4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Torres-Ruiz M, Robinson-Ector K, Attinson D, Trotter J, Anise A, Clauser S. 2018. A portfolio analysis of culturally tailored trials to address health and healthcare disparities. Int J Environ Res Public Health 15:1859. doi: 10.3390/ijerph15091859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sanders Thompson VL, Cavazos-Rehg PA, Jupka K, Caito N, Gratzke J, Tate KY, Deshpande A, Kreuter MW. 2008. Evidential preferences: cultural appropriateness strategies in health communications. Health Educ Res 23:549–559. doi: 10.1093/her/cym029. [DOI] [PubMed] [Google Scholar]
  • 214.Tapsell LC. 2017. Dietary behaviour changes to improve nutritional quality and health outcomes. Chronic Dis Transl Med 3:154–158. doi: 10.1016/j.cdtm.2017.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Wolf RL, Morawetz M, Lee AR, Koch PA, Contento IR, Zybert P, Green PHR, Lebwohl B. 2020. A cooking-based intervention promotes gluten-free diet adherence and quality of life for adults with celiac disease. Clin Gastroenterol Hepatol 18:2625–2627. doi: 10.1016/j.cgh.2019.09.019. [DOI] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES